10.08.2013 Views

Policies for Rare Diseases and Orphan Drugs - KCE

Policies for Rare Diseases and Orphan Drugs - KCE

Policies for Rare Diseases and Orphan Drugs - KCE

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>Policies</strong> <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong> <strong>and</strong> <strong>Orphan</strong><br />

<strong>Drugs</strong><br />

<strong>KCE</strong> reports 112C<br />

Federaal Kenniscentrum voor de Gezondheidszorg<br />

Centre fédéral d’expertise des soins de santé<br />

Belgian Health Care Knowledge Centre<br />

2009


The Belgian Health Care Knowledge Centre<br />

Introduction : The Belgian Health Care Knowledge Centre (<strong>KCE</strong>) is an organization<br />

of public interest, created on the 24 th of December 2002 under the<br />

supervision of the Minister of Public Health <strong>and</strong> Social Affairs.<br />

<strong>KCE</strong> is in charge of conducting studies that support the political<br />

decision making on health care <strong>and</strong> health insurance.<br />

Administrative Council<br />

Actual Members : Gillet Pierre (President), Cuypers Dirk (Deputy President),<br />

Avontroodt Yol<strong>and</strong>e, De Cock Jo (Deputy President), Baeyens Jean-<br />

Pierre, De Ridder Henri, de Stexhe Olivier, Godin Jean-Noël, Goyens<br />

Floris, Maes Jef, Mertens Pascal, Mertens Raf, Moens Marc, Perl<br />

François, Van Massenhove Frank (Deputy President), Degadt Peter,<br />

Verertbruggen Patrick, Schetgen Marco, Devos Daniël, Smeets Yves.<br />

Substitute Members : Cuypers Rita, Decoster Christiaan, Collin Benoit, Stamatakis Lambert,<br />

Vermeyen Karel, Kesteloot Katrien, Ooghe Bart, Lernoux Frederic,<br />

V<strong>and</strong>erstappen Anne, Palsterman Paul, Messiaen Geert, Remacle Anne,<br />

Lemye Rol<strong>and</strong>, Poncé Annick, Smiets Pierre, Bertels Jan, Lucet<br />

Catherine.<br />

Government commissioner: Roger Yves<br />

Management<br />

Chief Executive Officer a.i. : Jean-Pierre Closon<br />

In<strong>for</strong>mation<br />

Federaal Kenniscentrum voor de gezondheidszorg - Centre fédéral d’expertise des soins de santé –<br />

Belgian Health Care Knowlegde Centre.<br />

Centre Administratif Botanique, Doorbuilding (10th floor)<br />

Boulevard du Jardin Botanique 55<br />

B-1000 Brussels<br />

Belgium<br />

Tel: +32 [0]2 287 33 88<br />

Fax: +32 [0]2 287 33 85<br />

Email : info@kce.fgov.be<br />

Web : http://www.kce.fgov.be


<strong>Policies</strong> <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong> <strong>and</strong><br />

<strong>Orphan</strong> <strong>Drugs</strong><br />

<strong>KCE</strong> reports 112C<br />

ALAIN DENIS, STEVEN SIMOENS, CHRISTEL FOSTIER, LUT MERGAERT, IRINA CLEEMPUT<br />

Federaal Kenniscentrum voor de Gezondheidszorg<br />

Centre fédéral d’expertise des soins de santé<br />

Belgian Health Care Knowledge Centre<br />

2009


<strong>KCE</strong> reports 112C<br />

Title : <strong>Policies</strong> <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong> <strong>and</strong> <strong>Orphan</strong> <strong>Drugs</strong><br />

Authors : Alain Denis (Yellow Window Management Consultants), Steven Simoens<br />

(K.U.Leuven), Christel Fostier (Yellow Window Management<br />

Reviewers:<br />

Consultants), Lut Mergaert (Yellow Window Management Consultants),<br />

Irina Cleemput (<strong>KCE</strong>)<br />

Mattias Neyt (<strong>KCE</strong>), Frank Hulstaert (<strong>KCE</strong>)<br />

External experts : Jean-Jacques Cassiman (K.U.Leuven/U.Z. Leuven), Marc Dooms<br />

Acknowledgements<br />

(K.U.Leuven/U.Z. Leuven), François Eyskens (University of Antwerp),<br />

André Lhoir (FAGG/AFMPS), Philippe Van Wilder (RIZIV/INAMI)<br />

We would like to thank the EMEA <strong>and</strong> Hans-Georg Eichler in particular<br />

<strong>for</strong> the fruitful collaboration on part of this project <strong>and</strong> Frank Hulstaert<br />

<strong>for</strong> review of the EMEA files. The study team also wants to thank<br />

explicitly Marc Dooms <strong>for</strong> his valuable contribution <strong>and</strong> availability<br />

throughout the study period.<br />

External validators: Gepke Delwel (College voor Zorgverzekeringen (CVZ), the<br />

Conflict of interest:<br />

Netherl<strong>and</strong>s), Alastair Kent (Genetic Interest Group, UK), Marc Bogaert<br />

(U.Z. Gent)<br />

François Eyskens received a fee <strong>for</strong> participating in the International Fabry<br />

Expert Group which was logistically supported by Shire (October 2008)<br />

<strong>and</strong> was reimbursed by Shire as the organizer of the Fabry disease<br />

symposium in Mexico (October 2008). Marc Bogaert is chairman of the<br />

College <strong>for</strong> physicians <strong>for</strong> orphan drugs at the National Institute <strong>for</strong><br />

Health <strong>and</strong> Disability Insurance. Alastair Kent received reimbursement of<br />

expenses from Genzyme to support participation as a speaker at a<br />

conference <strong>and</strong> to chair a panel discussion at a conference.<br />

Disclaimer: The external experts were consulted about a (preliminary) version of the<br />

scientific report. Subsequently, a (final) version was submitted to the<br />

validators. The validation of the report results from a consensus or a<br />

voting process between the validators. Only the <strong>KCE</strong> is responsible <strong>for</strong><br />

errors or omissions that could persist. The policy recommendations are<br />

also under the full responsibility of the <strong>KCE</strong>.<br />

Layout: Ine Verhulst<br />

Brussels, 19 th February 2010 (2 nd print; 1 st print: 9 th July 2009)<br />

Study nr 2008-04<br />

Domain: Health Technology Assessment (HTA)<br />

MeSH: <strong>Rare</strong> <strong>Diseases</strong>; <strong>Orphan</strong> Drug Production; Health Policy; Legislation, Drug<br />

NLM classification : QZV 736<br />

Language: English<br />

Format: Adobe® PDF (A4)<br />

Legal depot: D/2009/10.273/32<br />

How to refer to this document?<br />

Denis A, Simoens S, Fostier C, Mergaert L, Cleemput I. <strong>Policies</strong> <strong>for</strong> <strong>Rare</strong> diseases <strong>and</strong> <strong>Orphan</strong> <strong>Drugs</strong>.<br />

Health Technology Assessment (HTA). Brussels: Belgian Health Care Knowledge Centre (<strong>KCE</strong>); 2009.<br />

<strong>KCE</strong> reports 112C (D/2009/10.273/32)


<strong>KCE</strong> reports 112C <strong>Orphan</strong> drugs i<br />

Executive summary<br />

BACKGROUND AND AIMS OF THE STUDY<br />

METHODS<br />

Due to low prevalence, rare diseases have traditionally been neglected by industry <strong>and</strong><br />

by the scientific, medical <strong>and</strong> political communities. It is estimated that there are<br />

currently between 5000 <strong>and</strong> 7000 different rare or rare diseases. Both in the United<br />

States (US) <strong>and</strong> in the European Union (EU), schemes were launched to stimulate the<br />

development of ‘orphan drugs’, drugs developed to treat these rare diseases. The<br />

rationale behind these legislations is to compensate industry <strong>for</strong> the risks <strong>and</strong> lower<br />

potential return on investment as a consequence of the inherently low number of<br />

patients. These schemes are considered as a success with an increasing number of<br />

<strong>Orphan</strong> Designation requests filed at the US Food <strong>and</strong> Drug Administration (FDA) <strong>and</strong><br />

the European Medicines Agency (EMEA). <strong>Orphan</strong> Designation refers to granting the<br />

orphan status to a medicinal product, while Marketing Authorisation refers to the<br />

approval to bring the product to the market. Since the development of the regulation to<br />

promote research <strong>and</strong> development on orphan drugs in the EU in 2000, 522 drugs<br />

obtained <strong>Orphan</strong> Designation <strong>and</strong> 47 received Marketing Authorisation.<br />

While <strong>Orphan</strong> Designation <strong>and</strong> Marketing Authorisation <strong>for</strong> these orphan drugs are<br />

decisions taken at the EU level, the drug reimbursement decision is a member state<br />

(MS) responsibility. National drug reimbursement committees (DRCs) are facing the<br />

trend of the increasing number of new orphan drugs entering the market <strong>and</strong> expect a<br />

relative increase in the budget spent on orphan drugs as compared to drugs <strong>for</strong> more<br />

common diseases mainly because of the high prices charged <strong>for</strong> these orphan drugs.<br />

This study’s objectives are to:<br />

1. provide an overview of the commonly used definitions <strong>for</strong> ‘rare diseases’ <strong>and</strong><br />

‘orphan drugs’ <strong>and</strong> describe the particularities of orphan drugs as compared<br />

to drugs <strong>for</strong> regular diseases;<br />

2. describe the regulatory processes <strong>for</strong> orphan drugs from <strong>Orphan</strong> Designation<br />

to reimbursement;<br />

3. compare the Belgian orphan drug reimbursement policy with other countries;<br />

4. estimate the current budget impact of orphan drugs <strong>and</strong> <strong>for</strong>ecast the<br />

expected future budget impact;<br />

5. <strong>for</strong>mulate recommendations <strong>for</strong> policy makers concerning orphan drugs.<br />

A narrative review, based on regulatory documents <strong>and</strong> published articles, provides an<br />

overview of the definitions <strong>for</strong> rare diseases <strong>and</strong> orphan drugs. For the description of<br />

regulatory processes <strong>for</strong> orphan drugs regulatory documents were examined <strong>and</strong><br />

interviews per<strong>for</strong>med with experts <strong>and</strong> key actors involved in the processes, both at<br />

national level <strong>and</strong> at European level (EMEA). Representatives of various stakeholders,<br />

including the pharmaceutical industry, patient organisations, policy makers, <strong>and</strong> experts<br />

involved in the assessment of orphan drug files were interviewed.<br />

To examine the consistency of the in<strong>for</strong>mation provided by industry to the different<br />

authorities at different levels <strong>and</strong> the extent to which in<strong>for</strong>mation available at European<br />

level could be used or be made more useful <strong>for</strong> national DRCs, a comparison was made<br />

between the clinical files submitted to the EMEA <strong>for</strong> obtaining marketing Authorisation,<br />

the resulting European Public Assessment Reports (EPARs) <strong>and</strong> the clinical evidence<br />

submitted to the Belgian National Institute <strong>for</strong> Health <strong>and</strong> Disability Insurance (NIHDI)<br />

as part of a drug reimbursement request. This was done <strong>for</strong> 15 specific drug-indication<br />

combinations. This collaboration between the EMEA <strong>and</strong> a national Health Technology<br />

Assessment (HTA) agency was unique <strong>and</strong> shows that it can serve a mutual interest,<br />

especially concerning orphan drugs.


ii <strong>Orphan</strong> drugs <strong>KCE</strong> reports 112C<br />

Additionally, a critical appraisal was made of the clinical <strong>and</strong> economic evidence<br />

submitted to the Belgian DRC <strong>for</strong> 8 cases. These were not necessarily included in the<br />

15 cases examined at the EMEA. The critical appraisal mainly looked at the type <strong>and</strong><br />

level of evidence submitted as well as the methodological st<strong>and</strong>ards applied to drug<br />

reimbursement files <strong>for</strong> orphan drugs.<br />

Six countries were included in the cross-country comparison of orphan drug<br />

reimbursement procedures: Belgium, France, Italy, the Netherl<strong>and</strong>s, Sweden <strong>and</strong> the<br />

United Kingdom. The country comparison relied on grey literature as well as on a<br />

survey among experts of the respective countries.<br />

For all reimbursed orphan drugs in Belgium the budget impact was estimated based on<br />

in<strong>for</strong>mation in the reimbursement request file <strong>and</strong> publicly available in<strong>for</strong>mation. In<br />

addition, simulations of the expected future budget impact of orphan drugs were made,<br />

using as a basis the average number of orphan drugs receiving marketing Authorisation<br />

each year, the proportion of orphan drugs obtaining reimbursement in Belgium <strong>and</strong> the<br />

average yearly orphan drug cost per patient.<br />

DEFINITIONS AND PARTICULARITIES OF ORPHAN DRUGS<br />

In the European legislation a rare disease is defined as a life-threatening or chronically<br />

debilitating condition with a prevalence of 5 patients per 10 000 people or less.<br />

However, a variety of definitions of rare diseases <strong>and</strong> of orphan drugs is used in the<br />

legislation of various countries.<br />

Also, a number of challenges exist with respect to the development, Marketing<br />

Authorisation, pricing, reimbursement <strong>and</strong> post-marketing follow-up of orphan drugs.<br />

Because of the low prevalence of these diseases, the development of a treatment is<br />

usually not considered economically interesting <strong>for</strong> a company. This situation might<br />

create unequal access to treatment between patients suffering from an rare disease <strong>and</strong><br />

patients suffering from a more common disease. There<strong>for</strong>e, the EU has created a series<br />

of incentives <strong>for</strong> the development of orphan drugs, such as fee reductions, protocol<br />

assistance <strong>and</strong> 10 years of market exclusivity.<br />

<strong>Orphan</strong> drugs are usually expensive. Considering all orphan drugs reimbursed in<br />

Belgium in 2008, the cost per patient per year was estimated to range from €6000 (<strong>for</strong><br />

the treatment of gastro-intestinal stromal tumour) to €312 000 (<strong>for</strong> the treatment of<br />

mucopolysaccharidosis type I). Companies justify these high prices by claiming that the<br />

high costs of research <strong>and</strong> development <strong>for</strong> orphan drugs can only be recouped from a<br />

small number of patients by asking higher prices. An additional explanation might be the<br />

monopoly situation that emerges when there are no alternative treatments yet <strong>for</strong> a<br />

specific rare condition <strong>and</strong> when, at the same time, no Marketing Authorisation can be<br />

granted to a similar product <strong>for</strong> the same therapeutic indication. It is worth noting that<br />

some products that were originally approved as orphan drugs <strong>and</strong> as such benefited<br />

from special measures later became top sellers, either because the indications were<br />

extended to more common diseases or because the rare indication became more<br />

frequent.


<strong>KCE</strong> reports 112C <strong>Orphan</strong> drugs iii<br />

ORPHAN DESIGNATION AND MARKETING<br />

AUTHORISATION<br />

To qualify <strong>for</strong> the special regulatory arrangements <strong>for</strong> orphan drugs, a drug has to<br />

obtain the orphan status through the <strong>Orphan</strong> Designation procedure at the EMEA.<br />

Afterwards, when the drug is ready <strong>for</strong> market introduction, Marketing Authorisation<br />

can be requested. At the EMEA two separate committees are responsible: the<br />

Committee <strong>for</strong> <strong>Orphan</strong> Medicinal Products (COMP) <strong>and</strong> the Committee <strong>for</strong> Human<br />

Medicinal Products (CHMP) respectively.<br />

<strong>Orphan</strong> Designation is subject to two conditions:<br />

• the medicinal product is intended <strong>for</strong> the diagnosis, prevention or<br />

treatment of a life-threatening or chronically debilitating condition that<br />

either affects less than 5 in 10 000 persons of the Community; or that<br />

without incentives is unlikely to generate sufficient return on investment<br />

to justify the expenditure;<br />

• there is an absence of solution or the drug brings a significant benefit<br />

compared to the present situation.<br />

The EMEA criteria used <strong>for</strong> <strong>Orphan</strong> Designation are different from the FDA’s. The FDA<br />

also considers the prevalence criterion, but more from the perspective of economic<br />

viability than from the perspective of existing alternatives. In the US, <strong>Orphan</strong><br />

Designation cannot be reconsidered, whereas it can in the EU. The FDA grants market<br />

exclusivity <strong>for</strong> 7 years.<br />

In contrast to many of the other drugs, where a national procedure still exists <strong>for</strong><br />

obtaining Marketing Authorisation, since 2005 orphan drugs can only obtain Marketing<br />

Authorisation through the centralized procedure at the EMEA. A Marketing<br />

Authorisation request is assessed by the CHMP supported by the preparatory work of<br />

the rapporteurs <strong>and</strong> their teams. The CHMP gives an opinion to the European<br />

Commission, which finally decides on the Marketing Authorisation. Based on the CHMP<br />

assessment report a European Public Assessment Report (EPAR) is prepared <strong>and</strong><br />

published on the web-site of the EMEA.<br />

A drug cannot receive Marketing Authorisation <strong>for</strong> an rare indication <strong>and</strong> a non-rare<br />

indication. In case of a conflict, the rare indication has to be dropped or marketing<br />

Authorisation will have to be requested <strong>for</strong> the drug under a different name.<br />

Consequently, identical products can be brought to the market under different names<br />

<strong>and</strong> prices.<br />

A company can decide to make a drug that has not yet obtained Marketing<br />

Authorisation but that is being considered by the EMEA, available on the market under<br />

the <strong>for</strong>mat of a ‘compassionate use programme’. The rules <strong>and</strong> conditions of the<br />

compassionate use programmes are organized either at the European level by the EMEA<br />

or at the national level by the individual member states.


iv <strong>Orphan</strong> drugs <strong>KCE</strong> reports 112C<br />

ORPHAN DRUG REIMBURSEMENT IN BELGIUM<br />

Drug reimbursement decisions are taken by the Minister of Social Affairs, after advice<br />

from the Drug Reimbursement Committee (DRC). <strong>Orphan</strong> drugs follow the same<br />

procedure as Class I pharmaceutical products, i.e. products <strong>for</strong> which the company<br />

claims a therapeutic added value. However, unlike <strong>for</strong> Class I pharmaceutical products,<br />

no pharmacoeconomic evaluation has to be submitted <strong>for</strong> orphan drugs. A decision on<br />

the reimbursement is taken within 180 days following the submission of the<br />

reimbursement request.<br />

At the end of December 2008, 31 orphan drugs were reimbursed in Belgium (including<br />

two products that do not have orphan drug status, but reimbursed <strong>for</strong> an rare<br />

indication) <strong>for</strong> a total of 35 rare indications. <strong>Orphan</strong> drugs are fully reimbursed.<br />

The prescription <strong>and</strong> individual reimbursement of orphan drugs is subject to conditions.<br />

The treating specialist physician must first obtain the approval of a Medical Advisor of<br />

the patient’s sickness fund to prescribe the medicine. The Medical Advisor can, but is<br />

not obliged to, request the advice of a “College of Medical Doctors <strong>for</strong> <strong>Orphan</strong> <strong>Drugs</strong>”<br />

(CMDOD). In practice, all sickness funds have agreed to refer all requests to the<br />

CMDOD if one exists. Separate Colleges exist <strong>for</strong> separate products. It is the DRC that<br />

decides whether or not a College is established. Individual reimbursement decisions are<br />

taken on a case by case basis by the CMDOD. At the end of 2008, there were 18<br />

colleges <strong>for</strong> 18 out of the 31 orphan drugs.<br />

If a medicinal product is not yet on the Belgian list of reimbursed pharmaceutical<br />

products, the patient may be able to benefit from a compassionate use or medical need<br />

programs by the company or in case the drug is already available on the market, request<br />

reimbursement through the Special Solidarity Fund (SSF). Conditions <strong>for</strong> compassionate<br />

use or reimbursement through the SSF are defined by law. In 2007, orphan drugs<br />

accounted <strong>for</strong> about 35% of the SSF’s total budget.<br />

INTERNATIONAL COMPARISON<br />

As in France, Italy <strong>and</strong> Sweden, Belgium has a number of centres of reference <strong>for</strong> rare<br />

diseases that are recognized by the NIHDI. Belgium has 8 centres <strong>for</strong> human genetics,<br />

10 centres <strong>for</strong> monogenic metabolic disorders <strong>and</strong> 6 centres <strong>for</strong> neuromuscular<br />

disorders, although these centres are not completely comparable to the centres of<br />

excellence in other countries. However, no <strong>for</strong>mal network of centres <strong>for</strong> rare diseases<br />

exists in this country. Centres of reference can build up expertise in specific rare<br />

diseases more easily than in cases where patients remain scattered over the country.<br />

In comparison with other countries, national measures to promote research on orphan<br />

drugs are relatively limited in Belgium. Sales of orphan drugs are exempted from taxes<br />

but no <strong>for</strong>mal research or support programmes are developed like in France, Italy <strong>and</strong><br />

the Netherl<strong>and</strong>s.<br />

Most countries, except <strong>for</strong> Sweden <strong>and</strong> the UK, compare the price requested by the<br />

company to the price in other countries. This creates incentives <strong>for</strong> the companies to<br />

introduce their products first in countries where a price requested as well as<br />

reimbursement are relatively easy to obtain compared to other countries. The UK has<br />

set up a system of profit control to control prices <strong>and</strong> Sweden uses a system of<br />

negotiation at the regional level.<br />

In Belgium <strong>and</strong> the UK, the distribution of orphan drugs occurs through hospital<br />

pharmacies only, although no <strong>for</strong>mal prohibition exists against the sales of orphan drugs<br />

in open pharmacies. In the other countries orphan drugs can also be delivered through<br />

community pharmacies. In Belgium, the UK <strong>and</strong> Italy prescription of orphan medication<br />

is the exclusive responsibility of a specialist physician. In all countries, specific conditions<br />

apply to the prescription of orphan drugs. An interesting case in this respect is Italy,<br />

where an orphan drug <strong>for</strong> an individual patient can only be delivered upon registration<br />

of the treatment start <strong>and</strong> follow-up in a national disease registry.


<strong>KCE</strong> reports 112C <strong>Orphan</strong> drugs v<br />

EVIDENCE LEVELS IN REIMBURSEMENT FILES<br />

To obtain reimbursement of an orphan drug, companies have to submit evidence on the<br />

efficacy <strong>and</strong> preferably also on the effectiveness of the drug as well as a budget impact<br />

analysis to the DRC.<br />

The evidence on clinical effectiveness is typically limited <strong>for</strong> orphan drugs, especially <strong>for</strong><br />

drugs targeting extremely small groups of patients. Due to the small number of patients<br />

clinical studies are rarely sufficiently powered to detect significant results on hard<br />

clinical endpoints. Moreover, the natural history of the disease is usually unknown, as<br />

physicians only have limited experience with the disease. The level of evidence in<br />

orphan drug reimbursement requests is there<strong>for</strong>e generally low. However, <strong>for</strong> nearly all<br />

products we examined at least one r<strong>and</strong>omized controlled trial (RCTs) was per<strong>for</strong>med<br />

demonstrating the possibility to per<strong>for</strong>m RCTs <strong>for</strong> orphan drugs.<br />

With respect to the choice of the comparator, it has been observed that alternative<br />

treatments were available <strong>for</strong> 15 orphan drugs, <strong>and</strong> that different orphan drugs<br />

sometimes have the same indication.<br />

Budget impact analyses in drug reimbursement files are often incomplete, in that they<br />

do not account <strong>for</strong> the budgetary impact of the product in the different indications <strong>for</strong><br />

which the product can be used. Companies can submit separate files <strong>for</strong> separate<br />

indications, in which case they calculate the budget impact of the indication <strong>for</strong> which<br />

the request is submitted, while the DRC should be able to consider the total budget<br />

impact of successive reimbursement files of an orphan drug relating to different<br />

indications. Methodological guidelines <strong>for</strong> budget impact analyses do not exist yet.<br />

CLINICAL INFORMATION AT DIFFERENT LEVELS<br />

The clinical file submitted to the EMEA in the context of a Marketing Authorisation<br />

request is not available <strong>for</strong> the DRCs of the member states, but a public assessment<br />

report (EPAR) is published <strong>for</strong> drugs that obtained Marketing Authorisation. The Belgian<br />

DRC asks companies to send the end-of-study reports as part of their drug<br />

reimbursement request file. Although companies are not obliged to do this, they usually<br />

provide these reports.<br />

The in<strong>for</strong>mation from the clinical files submitted to the EMEA is generally wellrepresented<br />

in the EPARs, although some suggestions could be made to further<br />

improve the utility of the EPARs <strong>for</strong> the national DRCs. For some drugs, a selective<br />

representation of the study results was found in the drug reimbursement request file:<br />

absence of the results of a negative study, of a phase I/II study <strong>and</strong> of an extension study<br />

<strong>and</strong> a vague description of the improvement without mentioning that these results were<br />

not significant from a statistical point of view. In 4 of the 15 examined cases more<br />

in<strong>for</strong>mation was provided in the NIHDI file than was available in the EMEA file, which<br />

can be explained by the time lag between the Marketing Authorisation request <strong>and</strong> the<br />

drug reimbursement request.<br />

Another observation is that an orphan drug may receive approval under exceptional<br />

circumstances based on surrogate endpoints only. In such cases the in<strong>for</strong>mation<br />

included in the EPAR, may not be relevant as such <strong>for</strong> the national decision maker, since<br />

the decision maker is mostly interested in clinically relevant outcomes parameters. In<br />

such cases the EMEA may request an additional RCT including these hard endpoints as a<br />

postmarketing requirement. Un<strong>for</strong>tunately, the results of such studies are not always<br />

made public by the EMEA <strong>and</strong> the EPAR is not automatically updated.


vi <strong>Orphan</strong> drugs <strong>KCE</strong> reports 112C<br />

BUDGET IMPACT ANALYSIS AND FORECAST<br />

The NIHDI expenditures on orphan drugs in Belgium is estimated to have been about<br />

€66 million or over 5 % of total hospital drug budget in 2008 <strong>and</strong> further estimates<br />

indicate the future cost will be well above 10 % of hospital drug budget in five years<br />

from now. <strong>Orphan</strong> drugs represent probably 2% of total drug reimbursement<br />

expenditures by NIHDI in 2009, <strong>and</strong> are estimated to represent close to 4% in 2013.<br />

This increasing cost creates an additional upward pressure on health care budgets <strong>and</strong><br />

may challenge the limits of solidarity between citizens.<br />

The high prices combined with the growing budget impact of orphan drugs also<br />

negatively affect the image of the orphan drugs among decision-makers.<br />

RECOMMENDATIONS<br />

EUROPEAN RECOMMENDATIONS<br />

Disease <strong>and</strong> patient registries<br />

• Priorities <strong>for</strong> research on rare diseases should be defined on European<br />

level in order to target public research funds <strong>for</strong> research <strong>and</strong><br />

development of orphan drugs.<br />

• For the high priority rare diseases, Europe should invest in setting up<br />

registries as early as possible; preferably be<strong>for</strong>e a drug is being developed<br />

<strong>for</strong> the disease. Data on the natural history of the disease <strong>and</strong> baseline<br />

risks are indispensable <strong>for</strong> describing the epidemiology of the disease <strong>and</strong><br />

put into context the clinical effectiveness of a treatment. The Directive<br />

95/46/EC of the European Parliament <strong>and</strong> of the Council of 24 October<br />

1995 on the protection of individuals with regard to the processing of<br />

personal data <strong>and</strong> on the free movement of such data should be respected<br />

when setting up these registries <strong>and</strong> using their data.<br />

• HTA agencies could play a role in the design of patient registries to<br />

ensure that the data collected can be used to help appreciate the<br />

effectiveness <strong>and</strong> cost-effectiveness obtained <strong>for</strong> novel drugs.<br />

• The aggregated data from the registries should be publicly available.<br />

• Funding <strong>and</strong> governance of the registries should be independent from the<br />

company developing an orphan drug. Innovative models combining<br />

European <strong>and</strong> national funds could be explored to set up such a system,<br />

the beneficiaries being the companies considering the clinical development<br />

of an orphan drug, the medicine agencies in assessing efficacy <strong>and</strong> safety,<br />

the national health care insurance funds, <strong>and</strong> the patients.


<strong>KCE</strong> reports 112C <strong>Orphan</strong> drugs vii<br />

<strong>Orphan</strong> drug designation <strong>and</strong> marketing authorisation<br />

• Evidence from R<strong>and</strong>omized Controlled Trials (RCTs) with clinically<br />

relevant endpoints should remain the st<strong>and</strong>ard <strong>for</strong> granting marketing<br />

authorization. The clinical endpoints should also have relevance <strong>for</strong><br />

national reimbursement decisions.<br />

• Surrogate outcome measures should only be used if there is a clear link<br />

with final endpoints or when final outcomes cannot be measured over an<br />

acceptable period of time.<br />

• HTA agencies may provide valuable input at the EMEA level <strong>for</strong> the<br />

definition of the endpoints needed <strong>and</strong> level of clinical improvement<br />

required in phase 3 studies, such that the product would qualify <strong>for</strong><br />

reimbursement.<br />

• The criteria <strong>for</strong> obtaining the orphan drug status <strong>and</strong> the associated<br />

incentive mechanisms may have to be revised in order to avoid artificial<br />

sub-setting.<br />

• The marketing exclusivity period should be revised when a product turns<br />

out to be profitable after a specific period of time, considering all<br />

indications of the drug. EU regulation should define what profitability<br />

means as well as the period of time over which profitability is assessed.<br />

• The European Public Assessment Reports (EPARs) should always<br />

o include a st<strong>and</strong>ard <strong>and</strong> complete tabular overview of clinical trials<br />

which were per<strong>for</strong>med or are still ongoing, mentioning the primary<br />

endpoints <strong>and</strong> the results <strong>for</strong> the pre-defined analyses, along with<br />

their level of significance<br />

o be updated whenever new evidence becomes available from<br />

studies requested by the EMEA in case of marketing Authorisation<br />

under exceptional circumstances.<br />

NATIONAL RECOMMENDATIONS<br />

Reimbursement policy<br />

• A drug reimbursement request file <strong>for</strong> an orphan drug should contain the<br />

same elements as a reimbursement request file <strong>for</strong> a class 1<br />

pharmaceutical product, more specifically:<br />

o At least efficacy, but preferably effectiveness, on a clinically relevant<br />

outcome should be demonstrated.<br />

o Cost-effectiveness ratios should be presented to show what<br />

society is paying <strong>for</strong> a Quality Adjusted Life Year (QALY) or life<br />

year gained.<br />

o Budget impact analysis should be based on epidemiological data<br />

from registries.<br />

o St<strong>and</strong>ardised cost in<strong>for</strong>mation submitted to the Federal Public<br />

Service Economy in the context of pricing. This st<strong>and</strong>ardisation has<br />

yet to be realised.


viii <strong>Orphan</strong> drugs <strong>KCE</strong> reports 112C<br />

Price setting <strong>and</strong> budget impact control<br />

Centralisation of Prescription requests<br />

• Price setting would ideally be dealt with at the European level or<br />

coordinated between member states.<br />

• In the meantime, NIHDI might consider different options to control the<br />

prices of orphan drugs:<br />

o Requiring a justification <strong>for</strong> the price based on detailed in<strong>for</strong>mation<br />

on the investments made <strong>and</strong> the potential return at a global level.<br />

This should be accompanied by a regular monitoring.<br />

o Risk-sharing between the drug company <strong>and</strong> the public payer,<br />

based on price <strong>for</strong> per<strong>for</strong>mance or conditional reimbursement.<br />

The minimally expected level of effectiveness on specific endpoints<br />

<strong>and</strong> the consequence of not realising the expected result, should<br />

be clearly specified when risk-sharing schemes are envisaged.<br />

• If a product is already reimbursed other indications, the budget impact of<br />

the product <strong>for</strong> those indications should also be reported.<br />

• All requests <strong>for</strong> the reimbursement of an orphan drug <strong>for</strong> an individual<br />

patient should be submitted directly to one central counter organised by<br />

the secretariat of the DRC at the NIHDI. This secretariat needs to be<br />

rein<strong>for</strong>ced if charged with this additional task.<br />

• Reimbursement of an orphan drug <strong>for</strong> a particular patient should be<br />

conditional upon the delivery of the st<strong>and</strong>ardised in<strong>for</strong>mation <strong>for</strong> the<br />

patient registry.<br />

• For each of the orphan drugs, the secretariat composes a College<br />

consisting of a representative from the secretariat <strong>and</strong> external experts<br />

that are chosen <strong>for</strong> their expertise in the rare disease. The secretariat<br />

delivers the prescription requests to the appropriate college.<br />

• The secretariat should make sure that the reimbursement criteria are<br />

consistently applied <strong>and</strong> should centralise the registration data of all<br />

reimbursement requests as well as the decisions of the Colleges. It should<br />

publish these data to increase the transparency of the system, con<strong>for</strong>m<br />

the privacy law.<br />

• The secretariat should also act as a coordination centre <strong>for</strong> rare diseases.<br />

It should be able to refer a physician who is faced with a patient with an<br />

rare disease to an appropriate expert or reference centre. A central<br />

website with appropriate in<strong>for</strong>mation <strong>and</strong> links <strong>for</strong> all rare diseases <strong>and</strong><br />

orphan drugs would be of use in this context.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 1<br />

Scientific Summary<br />

Table of contents<br />

GLOSSARY ................................................................................................................................. 4<br />

1 INTRODUCTION ............................................................................................................ 6<br />

1.1 OBJECTIVES OF THIS STUDY .................................................................................................................. 7<br />

1.2 GENERAL METHODOLOGY OF THE STUDY ................................................................................... 7<br />

2 ORPHAN, RARE AND NEGLECTED DISEASES AND DRUGS: DEFINITIONS<br />

AND PARTICULARITIES ............................................................................................... 9<br />

2.1 INTRODUCTION ........................................................................................................................................ 9<br />

2.2 METHODOLOGY ........................................................................................................................................ 9<br />

2.3 ORPHAN, RARE AND NEGLECTED DISEASES AND DRUGS ....................................................... 9<br />

2.4 ORPHAN DRUGS ...................................................................................................................................... 10<br />

2.4.1 Background ...................................................................................................................................... 10<br />

2.4.2 Definitions ........................................................................................................................................ 11<br />

2.4.3 Development ................................................................................................................................... 11<br />

2.4.4 Marketing Authorisation ............................................................................................................... 12<br />

2.4.5 Pricing ............................................................................................................................................... 13<br />

2.4.6 Health technology assessment <strong>and</strong> reimbursement ................................................................ 14<br />

2.5 CONCLUSIONS ......................................................................................................................................... 15<br />

3 POLICY DESCRIPTION ............................................................................................... 17<br />

3.1 INTRODUCTION ...................................................................................................................................... 17<br />

3.2 EMEA PROCESS: FROM ORPHAN DESIGNATION TO MARKETING AUTHORISATION . 17<br />

3.2.1 Presentation of EMEA ................................................................................................................... 17<br />

3.2.2 Applying <strong>for</strong> <strong>Orphan</strong> Designation ............................................................................................... 18<br />

3.2.3 Applying <strong>for</strong> Marketing Authorisation ....................................................................................... 22<br />

3.2.4 Compassionate use ........................................................................................................................ 28<br />

3.3 FDA PROCESS: FROM ORPHAN DESIGNATION TO MARKETING AUTHORISATION .... 28<br />

3.3.1 Presentation of the FDA ............................................................................................................... 28<br />

3.3.2 <strong>Orphan</strong> Drug Designation ............................................................................................................ 29<br />

3.3.3 Marketing approval ........................................................................................................................ 31<br />

3.3.4 Compassionate use ........................................................................................................................ 31<br />

3.4 EMEA-FDA COMPARISON ..................................................................................................................... 31<br />

4 INTERNATIONAL COMPARISON OF RARE DISEASE AND DRUG MARKETS<br />

IN EUROPE .................................................................................................................... 34<br />

4.1 INTRODUCTION ...................................................................................................................................... 34<br />

4.2 METHODOLOGY ...................................................................................................................................... 34<br />

4.3 BELGIUM ...................................................................................................................................................... 35<br />

4.3.1 Institutional context ....................................................................................................................... 35<br />

4.3.2 Marketing Authorisation ............................................................................................................... 35<br />

4.3.3 Reimbursement ............................................................................................................................... 36<br />

4.3.4 Pricing ............................................................................................................................................... 43<br />

4.3.5 Distribution ..................................................................................................................................... 43<br />

4.3.6 Prescribing ....................................................................................................................................... 43<br />

4.4 FRANCE ....................................................................................................................................................... 45<br />

4.4.1 Institutional context ....................................................................................................................... 45<br />

4.4.2 Marketing Authorisation ............................................................................................................... 46<br />

4.4.3 Reimbursement ............................................................................................................................... 46<br />

4.4.4 Pricing ............................................................................................................................................... 48<br />

4.4.5 Distribution ..................................................................................................................................... 48<br />

4.4.6 Prescribing ....................................................................................................................................... 49<br />

4.5 ITALY ............................................................................................................................................................ 49<br />

4.5.1 Institutional context ....................................................................................................................... 49<br />

4.5.2 Marketing Authorisation ............................................................................................................... 50


2 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

4.5.3 Reimbursement ............................................................................................................................... 50<br />

4.5.4 Pricing ............................................................................................................................................... 51<br />

4.5.5 Distribution ..................................................................................................................................... 52<br />

4.5.6 Prescribing ....................................................................................................................................... 52<br />

4.6 THE NETHERLANDS ................................................................................................................................ 53<br />

4.6.1 Institutional context ....................................................................................................................... 53<br />

4.6.2 Marketing Authorisation ............................................................................................................... 54<br />

4.6.3 Reimbursement ............................................................................................................................... 55<br />

4.6.4 Pricing ............................................................................................................................................... 58<br />

4.6.5 Distribution ..................................................................................................................................... 58<br />

4.6.6 Prescribing ....................................................................................................................................... 58<br />

4.7 SWEDEN ...................................................................................................................................................... 59<br />

4.7.1 Institutional context ....................................................................................................................... 59<br />

4.7.2 Marketing Authorisation ............................................................................................................... 59<br />

4.7.3 Reimbursement ............................................................................................................................... 60<br />

4.7.4 Pricing ............................................................................................................................................... 61<br />

4.7.5 Distribution ..................................................................................................................................... 61<br />

4.7.6 Prescribing ....................................................................................................................................... 61<br />

4.8 UNITED KINGDOM ................................................................................................................................. 62<br />

4.8.1 Institutional context ....................................................................................................................... 62<br />

4.8.2 Marketing Authorisation ............................................................................................................... 64<br />

4.8.3 Reimbursement ............................................................................................................................... 64<br />

4.8.4 Pricing ............................................................................................................................................... 65<br />

4.8.5 Distribution ..................................................................................................................................... 66<br />

4.8.6 Prescribing ....................................................................................................................................... 66<br />

4.9 COMPARATIVE ANALYSIS ..................................................................................................................... 66<br />

5 CRITICAL ASSESSMENT ............................................................................................. 70<br />

5.1 INTRODUCTION ...................................................................................................................................... 70<br />

5.2 METHODOLOGY ...................................................................................................................................... 70<br />

5.2.1 Qualitative overview ...................................................................................................................... 70<br />

5.2.2 In-depth analysis ............................................................................................................................. 71<br />

5.3 QUALITATIVE OVERVIEW OF ALL REIMBURSEMENT DOSSIERS ............................................. 72<br />

5.4 IN-DEPTH ANALYSIS OF 15 SELECTED REIMBURSEMENT DOSSIERS ..................................... 74<br />

5.4.1 Comparison of the evaluations by EMEA .................................................................................. 74<br />

5.4.2 Comparison of the studies mentioned in the NIHDI file, the EMEA file <strong>and</strong> EPAR ........ 75<br />

6 BUDGET IMPACT ANALYSIS .................................................................................... 77<br />

6.1 METHODOLOGY ...................................................................................................................................... 77<br />

6.2 BUDGET IMPACT IN BELGIUM AT THE END OF 2008 ................................................................ 77<br />

6.3 BUDGET IMPACT FORECAST ............................................................................................................... 81<br />

7 DISCUSSION AND CONCLUSIONS ......................................................................... 85<br />

7.1 ORPHAN DRUG DESIGNATION AS A TACTICAL STEP ............................................................. 85<br />

7.2 PREVALENCE VERSUS ECONOMIC MOTIVES ................................................................................. 85<br />

7.3 ASSESSING CLINICAL ADDED VALUE ............................................................................................... 86<br />

7.4 THE NEED FOR A RIGHT BALANCE BETWEEN ETHICAL AND ECONOMIC CONCERNS<br />

........................................................................................................................................................................ 87<br />

7.5 PRICING ....................................................................................................................................................... 88<br />

7.6 EXTENSION OF INDICATIONS ........................................................................................................... 91<br />

7.7 GROWTH OF THE BUDGET IMPACT OF ORPHAN DRUGS ..................................................... 92<br />

7.8 VARIATIONS IN ACCESS AND USE AMONG MEMBER STATES ............................................... 92<br />

7.9 AWARENESS RAISING ............................................................................................................................. 94<br />

7.10 COLLEGES AND CONTROL OF ELIGIBILITY .................................................................................. 94<br />

7.11 USE OF REGISTRIES .................................................................................................................................. 96<br />

8 APPENDICES ................................................................................................................. 99


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 3<br />

8.1 OVERVIEW REIMBURSED ORPHAN DRUGS IN BELGIUM ........................................................ 100<br />

8.2 CHARACTERISTICS OF ORPHAN DRUGS SUBMITTED FOR REIMBURSEMENT IN<br />

BELGIUM, 2004-2008 .............................................................................................................................. 102<br />

8.3 QUALITATIVE QUESTIONNAIRE BENCHMARKING .................................................................. 104<br />

8.4 QUALITATIVE QUESTIONNAIRE PHARMACEUTICAL INDUSTRIES ..................................... 108<br />

8.5 LIST OF EXPERTS AND STAKEHOLDERS CONSULTED FOR THE STUDY ......................... 110<br />

8.5.1 List of interview respondents .................................................................................................... 110<br />

8.5.2 Consultations ................................................................................................................................ 110<br />

8.5.3 National experts ........................................................................................................................... 110<br />

8.6 LIST OF 14 ORPHAN DRUGS USED FOR EMEA – NIHDI COMPARISON ............................ 111<br />

9 REFERENCES ............................................................................................................... 112


4 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

GLOSSARY<br />

Afssaps Agence Française de Sécurité Sanitaire des Produits<br />

de Santé<br />

http://agmed.sante.gouv.fr/<br />

AIFA Agenzia Italiana del Farmaco (Italian Medicines<br />

Agency)<br />

http://www.agenziafarmaco.it<br />

ASMR Amélioration du Service medical rendu<br />

(Improvement in clinical added value)<br />

ATC Anatomical Therapeutic Chemical-code<br />

ATU Authorisation <strong>for</strong> Temporary Usage<br />

AWMSG All Wales Medicines Strategy Group http://www.wales.nhs.uk<br />

BNF British National Formulary<br />

CBG College ter Beoordeling van Geneesmiddelen (Dutch<br />

Medicines Evaluation Board)<br />

http://www.cbg-meb.nl/cbg/nl<br />

CEPS Comité Economique des Produits de Santé (French<br />

Healthcare Products Economic Committee)<br />

http://www.sante.gouv.fr/ceps/<br />

CHMP EMEA Committee <strong>for</strong> Medicinal Products <strong>for</strong> Human http://www.emea.europa.eu/htms/gen<br />

Use<br />

eral/contacts/CHMP/CHMP.html<br />

CMDOD Belgian College of Medical Doctors <strong>for</strong> <strong>Orphan</strong><br />

<strong>Drugs</strong> (College van Geneesheren voor<br />

Weesgeneesmiddelen / Collège de médecins pour<br />

des médicaments orphelins)<br />

COMP EMEA Committee on <strong>Orphan</strong> Medicinal Products http://www.emea.europa.eu/htms/gen<br />

eral/contacts/COMP/COMP.html<br />

CPA Dutch Committee <strong>for</strong> Pharmaceutical Aid<br />

DPBB Swedish Dental <strong>and</strong> Pharmaceutical Benefits Board http://www.tlv.se<br />

DRC Belgian Drug Reimbursement Commission<br />

(Commissie Tegemoetkoming Geneesmiddelen /<br />

Commission de Remboursement des Médicaments)<br />

DTC Diagnosis <strong>and</strong> Treatment Combinations (the<br />

Netherl<strong>and</strong>s)<br />

EC European Commission http://ec.europa.eu/<br />

EGAN European Genetic Alliance Network http://www.egan.eu/<br />

EMEA European Medicines Agency http://www.emea.europa.eu/<br />

EPAR European Public Assessment Report<br />

EU European Union http://europa.eu/<br />

Eurordis European Organisation <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong> http://www.eurordis.org<br />

FDA US Food <strong>and</strong> Drug Administration http://www.fda.gov/<br />

FPS Federal Public Service (<strong>for</strong>mer Belgian Ministry)<br />

GIS Groupe d’intérêt scientifique<br />

GVS Geneesmiddelenvergoedingssysteem (Dutch<br />

Medicines reimbursement system)<br />

HAS Haute Autorité de Santé (French High Health<br />

Authority)<br />

http://www.has-sante.fr<br />

HCIB Dutch Health Care Insurance Board (College voor<br />

Zorgverzekeringen)<br />

http://www.cvz.nl/<br />

HTA Health technology assessment<br />

ICER Incremental cost-effectiveness ratio<br />

INAHTA International Network of Agencies <strong>for</strong> Health<br />

Technology Assessment<br />

www.inahta.org<br />

MA Marketing Authorisation<br />

MAH Marketing Authorisation Holder<br />

MD Medical Doctor<br />

MHRA British Medicines <strong>and</strong> Healthcare products<br />

Regulatory Agency<br />

http://www.mhra.gov.uk/<br />

MS (European Union) Member States


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 5<br />

NCG National Commissioning Group (body of the British<br />

NHS)<br />

http://www.ncg.nhs.uk/<br />

NHS National Health Service<br />

NICE National Institute <strong>for</strong> Health <strong>and</strong> Clinical Excellence<br />

(UK)<br />

www.nice.org.uk<br />

NIHDI Belgian National Institute <strong>for</strong> Health <strong>and</strong> Disability<br />

Insurance (Rijksinstituut voor ziekte- en<br />

invaliditeitsverzekering / Institut National<br />

d’Assurance Maladie et d’Invalidité)<br />

www.NIHDI.be<br />

NORD National Organization <strong>for</strong> <strong>Rare</strong> Disorders (US) www.rarediseases.org<br />

OD <strong>Orphan</strong> drug<br />

ODD <strong>Orphan</strong> Drug Designation<br />

OOPD Office of <strong>Orphan</strong> Products Development of the FDA http://www.fda.gov/orphan/<br />

ORPHANET The portal <strong>for</strong> rare diseases <strong>and</strong> orphan drugs www.orpha.net<br />

PA Protocol assistance (EMEA) http://ec.europa.eu/enterprise/pharma<br />

ceuticals/orphanmp/index.htm<br />

PCT Primary Care Trust (United Kingdom)<br />

PPRS British Pharmaceutical Price Regulation Scheme<br />

QALY Quality Adjusted Life Year<br />

RCT R<strong>and</strong>omized controlled trial<br />

RoI Return on Investment<br />

SA Scientific advice http://ec.europa.eu/enterprise/pharma<br />

ceuticals/orphanmp/index.htm<br />

SAWP Scientific Advice Working Party of the EMEA<br />

SMC Scottish Medicines Consortium http://www.scottishmedicines.org.uk<br />

SMR Service Medical Rendu (clinical added value)<br />

SSF Belgian Special Solidarity Funds (Bijzonder<br />

http://www.NIHDI.fgov.be/care/nl/info<br />

Solidariteitsfonds / Fonds Spécial de Solidarité ) s/solidarity/index.htm<br />

WGO Stuurgroep Weesgeneesmiddelen (Dutch Steering<br />

Committee on <strong>Orphan</strong> <strong>Drugs</strong>)<br />

www.weesgeneesmiddelen.nl<br />

WHO World Health Organisation www.who.org<br />

ZonMw Dutch Organisation <strong>for</strong> Health Research <strong>and</strong><br />

Development<br />

http://www.zonmw.nl/


6 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

1 INTRODUCTION<br />

An rare disease is generally defined as a disease with a very low prevalence. Different<br />

operational definitions <strong>for</strong> rare diseases are used in legal documents <strong>and</strong> in literature. As<br />

such, the definition used in Europe differs from the one used in the United States.<br />

<strong>Rare</strong> diseases are often difficult to diagnose <strong>and</strong> specialized clinicians are most often<br />

scarce. Moreover, (drug) treatments <strong>for</strong> rare diseases are less likely to be produced by<br />

private companies because the market is too small <strong>and</strong> research <strong>and</strong> development costs<br />

<strong>for</strong> orphan products are usually too high to make the products profitable. <strong>Drugs</strong> used<br />

<strong>for</strong> the treatment of a rare disease are hereafter called orphan drugs.<br />

Both in the US <strong>and</strong> in the European Union incentives have been created to promote<br />

research <strong>and</strong> development on orphan drugs. Between 2000 <strong>and</strong> 2008 more than 590<br />

medicinal products received European orphan drug status. Almost 50 received<br />

marketing Authorisation in this period. About 30% of these were in the field of<br />

oncology <strong>and</strong> 27% in the field of endocrinology <strong>and</strong> metabolic disorders.<br />

It is estimated that there are currently between 5 000 <strong>and</strong> 8 000 different diseases that<br />

can be classified as rare. With less than 50 orphan drugs on the market at the end of<br />

2008, only a small part of the need <strong>for</strong> treatment of rare diseases is covered.<br />

Given the increasing number of orphan drugs <strong>and</strong> the high costs of orphan drugs,<br />

budgets spent to orphan drugs continue to increase. While in absolute numbers the<br />

total budget impact of orphan drugs might still be limited (about 2% of total hospital<br />

drug expenditures in 2009), their relative budget impact becomes steadily more<br />

important.<br />

As reimbursement policies with respect to orphan drugs differ between countries,<br />

access to orphan drugs also differs between countries. In the Belgian context,<br />

reimbursement of a product in Class I a requires evidence of the added therapeutic value<br />

of the product. However, due to the small number of patients with an rare disease, the<br />

clinical evidence base will often be weaker <strong>for</strong> orphan drug than <strong>for</strong> regular drugs.<br />

Economic evaluations of orphan drugs are often hampered by the limited evidence on<br />

clinical effectiveness <strong>for</strong> the drug. Moreover, using traditional approaches economic<br />

evaluations will usually find that orphan <strong>Drugs</strong> are not cost-effective because the cost<br />

<strong>for</strong> the additional health benefit the orphan drug treatment offers is usually high<br />

compared to many non)orphan treatments. 2<br />

The specific features of rare diseases <strong>and</strong> orphan drugs combined with the increasing<br />

number of rare diseases, makes them an issue of high priority <strong>for</strong> policy makers. On the<br />

one h<strong>and</strong> policy makers are faced with an increasing proportion of the health care<br />

budget being spent on orphan drugs, on the other h<strong>and</strong> they have to recognize the<br />

ethical <strong>and</strong> social dimension of rare disease treatment <strong>and</strong> deal with these under the<br />

constraint of not being able to expect the same level of clinical evidence <strong>for</strong> orphan<br />

drugs as <strong>for</strong> other drugs.<br />

a<br />

There are three added value classes in Belgium – the therapeutic value of a medicinal product is decided by<br />

the DRC <strong>and</strong> expressed in an added value class<br />

Class 1: medicinal products of which the therapeutic added value has been proven compared to existing<br />

therapeutic alternatives<br />

Class 2: medicinal products with no proven therapeutic added value compared to existing therapeutic<br />

alternatives<br />

Class 3: other medicinal products – categorized according to legislation<br />

Source: art. 5 Royal Decree 21/12/2001


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 7<br />

1.1 OBJECTIVES OF THIS STUDY<br />

The main objectives of this study are:<br />

1. to provide an overview of the commonly used definitions <strong>for</strong> ‘rare diseases’<br />

<strong>and</strong> ‘orphan drugs’ <strong>and</strong> describe the particularities of orphan drugs compared<br />

to regular drugs;<br />

2. to describe the regulatory process followed by an orphan drug, from orphan<br />

designation at the European level to reimbursement in Belgium <strong>and</strong> examine<br />

to what extent the in<strong>for</strong>mation produced by the authorities responsible <strong>for</strong><br />

orphan designation <strong>and</strong> Marketing Authorisation is directly useful <strong>for</strong> the drug<br />

reimbursement decision process;<br />

3. to compare the Belgian policy with regard to the reimbursement of orphan<br />

drugs with the procedures that exist in other countries <strong>for</strong> decision-making<br />

about the reimbursement of orphan drugs;<br />

4. to estimate the current budget impact of orphan drugs <strong>and</strong> make a prudent<br />

<strong>for</strong>ecast of the expected budget impact in the years to come, <strong>and</strong><br />

5. to <strong>for</strong>mulate recommendations <strong>for</strong> policy makers concerning orphan drugs.<br />

Chapter 2 gives an overview of the different definitions <strong>for</strong> rare diseases <strong>and</strong> orphan<br />

drugs. Chapter 3 describes the European process from orphan designation to Marketing<br />

Authorisation <strong>and</strong> compares this with the process in the US. Chapter 4 compares the<br />

orphan drug reimbursement policies of 6 countries, including Belgium. Chapter 5<br />

describes the extent to which the in<strong>for</strong>mation provided by the pharmaceutical<br />

companies to EMEA in order to obtain Marketing Authorisation <strong>and</strong> the public<br />

assessment report produced by EMEA corresponds with the in<strong>for</strong>mation available to<br />

the Belgian drug reimbursement committee (DRC) at the time drug reimbursement is<br />

requested. Chapter 6 includes an analysis of the current budget impact of orphan drugs<br />

in Belgium <strong>and</strong> makes a prudent <strong>for</strong>ecast of the expected budget impact in the coming<br />

years. Chapter 7 contains a discussion of the issues related to orphan drugs <strong>and</strong> chapter<br />

8 concludes the report with a number of recommendations <strong>for</strong> European <strong>and</strong> Belgian<br />

policy makers. We recommend to readers who want a quick insight to read the<br />

executive summary <strong>and</strong> chapter 7.<br />

1.2 GENERAL METHODOLOGY OF THE STUDY<br />

The methodology followed <strong>for</strong> this project can be summarised into seven activities.<br />

Activity 1: Desk research<br />

As a first activity, the contextual situation of the policy with regard to rare diseases <strong>and</strong><br />

orphan drugs in Belgium was analysed against the European background. This included a<br />

collection <strong>and</strong> review of relevant documents <strong>and</strong> scholarly publications relating to the<br />

particularities of orphan drugs (such as market access, pricing, patient care, health<br />

technology assessments …).<br />

Activity 2: Policy description of the processes at EMEA <strong>and</strong> FDA<br />

The aim was to compare the <strong>Orphan</strong> Designation <strong>and</strong> Marketing Authorisation<br />

processes of the EMEA <strong>and</strong> FDA. In<strong>for</strong>mation was collected through desk research <strong>and</strong><br />

interviews.<br />

Activity 3: Comparative analysis of the Belgian situation <strong>and</strong> of five other EU<br />

countries<br />

The third activity focussed on the Belgian reimbursement procedure: a description is<br />

provided of the criteria used <strong>for</strong> reimbursement of orphan drugs <strong>and</strong> of the differences<br />

of the decision process compared to other medicinal products. This work is based on<br />

qualitative research <strong>and</strong> interviews. The description of the Belgian situation is followed<br />

by an overview of the reimbursement procedure in France, Italy, the Netherl<strong>and</strong>s,<br />

Sweden <strong>and</strong> the United Kingdom. In<strong>for</strong>mation <strong>for</strong> this activity was collected through<br />

desk research <strong>and</strong> a survey based on a qualitative questionnaire.


8 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Activity 4: Budget impact analysis<br />

For all reimbursed orphan drugs in Belgium the budget impact was estimated based on<br />

the budget impact analysis presented by the companies in reimbursement request files<br />

<strong>and</strong> publicly available in<strong>for</strong>mation (analysis of actual <strong>and</strong> expected budget impact over<br />

the years). Also <strong>for</strong>ecasts <strong>and</strong> simulations of expected future budget impact of orphan<br />

drugs were made, using as a basis the average number of drugs getting marketing<br />

authorisation each year, the percentage of orphan drugs obtaining reimbursement in<br />

Belgium <strong>and</strong> the average cost per patient per year of orphan drugs.<br />

Activity 5: Critical assessment<br />

The critical assessment consisted of a ‘quick scan’ <strong>for</strong> all reimbursed orphan drugs in<br />

Belgium <strong>and</strong> a more in depth critical appraisal of eight cases (eight reimbursed <strong>and</strong> one<br />

negative case). The quick scan looked at the clinical <strong>and</strong> economic evidence provided in<br />

the context of the registration <strong>and</strong> reimbursement request files submitted to the<br />

NIHDI, whereas the in depth critical appraisal took into account the methodological<br />

st<strong>and</strong>ards of registration <strong>and</strong> reimbursement request files. The eight cases were<br />

selected according to a number of selection criteria (defined by the experts).<br />

Activity 6: Discussion of issues<br />

In Chapter 7 of this report eleven “issues” are presented which were identified through<br />

the study <strong>and</strong> which deserve attention at the policy-making level. This discussion offers<br />

some considerations which may serve as input <strong>for</strong> recommendations that follow from<br />

this study.<br />

In addition, personal interviews complemented the various other techniques used <strong>for</strong><br />

the activities described above. These interviews took place with key actors involved in<br />

the process, both at the national <strong>and</strong> at the EU level, as well as with representatives of<br />

the various stakeholders, from COMP members, over EMEA or NIHDI to patient<br />

organisations <strong>and</strong> the pharmaceutical industry.<br />

Activity 7: Recommendations<br />

The recommendations were written by the <strong>KCE</strong> based on the results of the scientific<br />

review.<br />

Important comment <strong>for</strong> the reader: most figures mentioned in this report in relation to<br />

the number of orphan drugs are based on the situation as of the 31 st of December<br />

2008.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 9<br />

2 ORPHAN, RARE AND NEGLECTED<br />

DISEASES AND DRUGS: DEFINITIONS AND<br />

PARTICULARITIES<br />

2.1 INTRODUCTION<br />

Some conceptual confusion exists around the terms ‘rare diseases’ <strong>and</strong> ‘orphan drugs’.<br />

Different definitions of rare diseases <strong>and</strong> of orphan drugs are used in existing legislation<br />

of various countries <strong>and</strong> in literature on the subject. This chapter gives an overview of<br />

commonly used definitions. <strong>Orphan</strong> drugs are distinct from common drugs in terms of<br />

their development, Marketing Authorisation (MA), pricing, reimbursement <strong>and</strong> postmarketing<br />

follow-up. This chapter also discusses economic challenges of developing <strong>and</strong><br />

marketing orphan drugs.<br />

2.2 METHODOLOGY<br />

A review of the international literature was carried out by searching the following<br />

electronic databases up to November 2008: PubMed, EMBASE, Bath In<strong>for</strong>mation <strong>and</strong><br />

Data Services, Cochrane Library, EconLit, <strong>and</strong> Social Science <strong>and</strong> Citation Index. Search<br />

terms included ‘orphan diseases’, ‘rare diseases’, ‘neglected diseases’, ‘orphan drugs’,<br />

‘ultra-orphan drugs’, ‘research <strong>and</strong> development’, ‘Marketing Authorisation’, ‘pricing’,<br />

‘reimbursement’, ‘health technology assessment’, ‘economic evaluation’, ‘costeffectiveness’,<br />

‘post-marketing follow-up’, ‘risk sharing’, ‘patient registry’, ‘access’ <strong>and</strong><br />

‘equity’. Additionally, the bibliography of included studies was checked <strong>for</strong> other<br />

relevant studies. Finally, in<strong>for</strong>mation about regulation with respect to rare diseases <strong>and</strong><br />

orphan drugs was gained from documents setting out international/national legislation.<br />

2.3 ORPHAN, RARE AND NEGLECTED DISEASES AND DRUGS<br />

The terms ‘orphan disease’ <strong>and</strong> ‘rare disease’ are frequently used interchangeably <strong>for</strong> a<br />

disease that affects only few persons in the population. However, according to some<br />

definitions orphan diseases comprise rare diseases as well as ‘neglected diseases’ 3 . The<br />

latter group consists of conditions that are prevalent in developing countries which are<br />

too poor to pay drug prices that render the new drug profitable <strong>for</strong> the patent-holding<br />

manufacturer. 4 This study will only focus on the group of rare diseases.<br />

When is a disease rare? The definitions that are used vary, but are usually expressed in<br />

prevalence figures. Table 1 gives an overview of the number of patients per 100 000<br />

individuals that countries apply to define a rare disease.<br />

According to the definition put <strong>for</strong>ward by the European Union (EU), rare diseases are<br />

life-threatening or chronically debilitating conditions with a prevalence of 50 out of<br />

100,000 or less 5 . According to the World Health Organisation, a rare disease affects at<br />

most 65 out of every 100,000 individuals. 3 Australia, Japan <strong>and</strong> the United States have<br />

set prevalences of 11 6 , 40 7 <strong>and</strong> 66 8 per 100,000 individuals respectively <strong>for</strong> a given rare<br />

disease. The Swedish National Board of Health <strong>and</strong> Welfare defines rare diseases as<br />

disorders or injuries that result in extensive h<strong>and</strong>icaps <strong>and</strong> that affect no more than 10<br />

per 100,000 individuals. 9


10 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Table 1: Definitions of rare diseases based on prevalence<br />

Country <strong>Rare</strong> diseases<br />

Prevalence on 100,000<br />

Source<br />

US 66 a<br />

<strong>Orphan</strong> Drug Act 1983<br />

EU 50 Regulation EC n° 141/2000<br />

Japan 40 <strong>Orphan</strong> Drug Act 1993<br />

Australia 11 <strong>Orphan</strong> Drug Program 1997<br />

SE 10 Swedish National Board of Health <strong>and</strong> Welfare<br />

FR 50 Regulation EC n° 141/2000<br />

NL 50 Regulation EC n° 141/2000<br />

WHO 65 WHO<br />

a Based on a total US population of 304,354,998 on 16 June 2008. Source: US Census Bureau<br />

(http://www.census.gov/). In scholarly literature, the US prevalence rates <strong>for</strong> <strong>Orphan</strong> Designation<br />

expressed per 10,000 inhabitants vary from less than 6 to ‘about 10’ though.<br />

Within the group of rare diseases, some diseases are relatively more common than<br />

others. As a result, a distinction is sometimes made between rare diseases <strong>and</strong> ultrarare<br />

diseases. Ultra-rare diseases are generally defined as affecting less than 10,000<br />

individuals on a population of 300 million individuals. 10 In the UK, the National Institute<br />

<strong>for</strong> Health <strong>and</strong> Clinical Excellence (NICE) sets the prevalence of an ultra-rare disease at<br />

less than 2 per 100,000 individuals. 11<br />

Regardless of the country-specific definition, it is estimated that between 5,000 <strong>and</strong><br />

8,000 distinct rare diseases exist today, 80% of which have identified genetic origins.<br />

Other rare diseases are the result of bacterial or viral infections <strong>and</strong> allergies, or are<br />

due to degenerative <strong>and</strong> proliferative causes. Together, rare diseases affect an important<br />

part of the population, estimated to be about 6% - 8% of the population of the<br />

European Union (EU), equivalent to 27-36 million people. 12<br />

2.4 ORPHAN DRUGS<br />

2.4.1 Background<br />

Due to their relatively low prevalence, rare diseases as a whole have traditionally been<br />

neglected by large parts of the scientific, medical <strong>and</strong> political communities. 13 With<br />

knowledge <strong>and</strong> awareness of the majority of rare diseases being scant or even absent,<br />

delay in diagnoses, lack of relevant in<strong>for</strong>mation <strong>and</strong> difficulty in finding specialised<br />

physicians are common problems <strong>for</strong> affected patients. While many patients even<br />

remain completely undiagnosed, even when recognised, thous<strong>and</strong>s of rare diseases<br />

cannot be treated because no therapies or drugs exist <strong>for</strong> them. This is primarily due to<br />

the fact that pharmaceutical companies are more interested in developing drugs <strong>for</strong><br />

common disorders that affect millions of people than in the treatments <strong>for</strong> a few<br />

14 2005<br />

<strong>and</strong> because of a scientific deficit as research is less oriented towards rare diseases. As a<br />

consequence, sufferers from rare diseases are not only disadvantaged in terms of<br />

likeliness <strong>and</strong> timeliness of being diagnosed as such, but are on top of that experiencing<br />

unequal access to therapy <strong>and</strong> treatment in comparison to patients suffering from<br />

‘common’ diseases. 15 In the past decades, this unequitable situation gained recognition as<br />

a serious public health problem <strong>and</strong> it became clear that the development of drugs <strong>for</strong><br />

rare diseases required special encouragement.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 11<br />

2.4.2 Definitions<br />

<strong>Orphan</strong> drugs are considered differently from other types of drugs by regulatory<br />

authorities. At EU level, discussions on orphan drugs started in the late nineties <strong>and</strong> led<br />

to the adoption of Regulation (EC) No 141/2000 of the European Parliament <strong>and</strong> of the<br />

Council of 16 December 1999, in which the justification <strong>for</strong> treating orphan drugs<br />

differently has been <strong>for</strong>mulated as follows (preamble, paragraphs 1 <strong>and</strong> 2): 5<br />

1. (Whereas…) some conditions occur so infrequently that the cost of developing <strong>and</strong><br />

bringing to the market a medicinal product to diagnose, prevent or treat the<br />

condition would not be recovered by the expected sales of the medicinal product;<br />

the pharmaceutical industry would be unwilling to develop the medicinal product<br />

under normal market conditions; these medicinal products are called ‘orphan’;<br />

2. (Whereas…) patients suffering from rare conditions should be entitled to the same<br />

quality of treatment as other patients; it is there<strong>for</strong>e necessary to stimulate the<br />

research, development <strong>and</strong> bringing to the market of appropriate medications by<br />

the pharmaceutical industry;<br />

Defining an ‘orphan drug’, Regulation (EC) No 141/2000 5 states (in Article 3.1) that:<br />

A medicinal product shall be designated as an orphan medicinal product if its sponsor can<br />

establish:<br />

(a) that it is intended <strong>for</strong> the diagnosis, prevention or treatment of a life-threatening or<br />

chronically debilitating condition affecting not more than five in 10 thous<strong>and</strong> persons in the<br />

Community when the application is made, or<br />

That it is intended <strong>for</strong> the diagnosis, prevention or treatment of a life-threatening, seriously<br />

debilitating or serious <strong>and</strong> chronic condition in the Community <strong>and</strong> that without incentives it is<br />

unlikely that the marketing of the medicinal product in the Community would generate<br />

sufficient return to justify the necessary investment;<br />

<strong>and</strong><br />

(b) that there exists no satisfactory method of diagnosis, prevention or treatment of the<br />

condition in question that has been authorised in the Community or, if such method exists, that<br />

the medicinal product will be of significant benefit to those affected by that condition.<br />

In summary, the arguments put <strong>for</strong>ward <strong>for</strong> considering orphan drugs differently than<br />

other drugs in the EU lie in the orphan drugs being economically not viable under<br />

normal market conditions <strong>and</strong> considerations of patient equity.<br />

2.4.3 Development<br />

In the EU, companies with an <strong>Orphan</strong> Designation <strong>for</strong> a medicinal product benefit from<br />

incentives such as: 16<br />

• protocol assistance (scientific advice during the product development<br />

phase);<br />

• direct access to the European Medicines Evaluation Agency (EMEA)<br />

Centralised Procedure with respect to registration;<br />

• Marketing Authorisation (10-year marketing exclusivity);<br />

• financial incentives (fee reduction b or exemptions, possible assistance with<br />

research <strong>and</strong> development) c ;<br />

• national incentives (detailed in an inventory of incentives made available by<br />

the European Commission 17 ).<br />

b Including a 100 % fee reduction <strong>for</strong> protocol assistance <strong>and</strong> 50% reduction <strong>for</strong> the application<br />

<strong>for</strong> Marketing Authorisation <strong>and</strong> 100% fee reduction <strong>for</strong> pre-authorisation inspections<br />

c In 2007, the funds made available by the Community <strong>for</strong> fee exemptions <strong>for</strong> orphan medicinal<br />

products amounted to € 6,000,000 (EMEA, 2007).


12 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Article 9 of Regulation (EC) No 141/2000 requires Member States to communicate to<br />

the Commission detailed in<strong>for</strong>mation concerning any measure they have enacted to<br />

support research into, <strong>and</strong> the development <strong>and</strong> availability of, orphan medicinal<br />

products or medicinal products that may be designated as such. The European<br />

Commission regularly publishes an inventory of such measures taken by the Member<br />

States according to article 9. d<br />

In the United States, incentives <strong>for</strong> the development of orphan medicinal products have<br />

been available since 1983. The United States’ <strong>Orphan</strong> Drug Act (ODA) 8 provides<br />

significant incentives <strong>for</strong> sponsors to develop <strong>and</strong> bring to the market drugs <strong>and</strong><br />

biologicals, including vaccines <strong>and</strong> in vivo diagnostics to tackle rare diseases 13 . These<br />

benefits include expedited review by the US Food <strong>and</strong> Drug Administration (FDA) <strong>and</strong><br />

thus shorter approval time, tax credits, seven years of marketing exclusivity <strong>and</strong><br />

reductions of certain fees. Marketing exclusivity means that similar products have no<br />

access to the market <strong>for</strong> seven years. Furthermore, research grants are available to<br />

support clinical trials of orphan drugs. To qualify <strong>for</strong> incentives, drugs must receive the<br />

‘<strong>Orphan</strong> Designation’ from the FDA’s Office of <strong>Orphan</strong> Products Development<br />

(OOPD), <strong>and</strong> then go through the normal evaluation process <strong>for</strong> safety <strong>and</strong> efficacy. 18<br />

The ODA specifies, next to the prevalence criterion, that a drug is also considered as<br />

an orphan drug if scientists <strong>and</strong> economists at the Food <strong>and</strong> Drug Administration (FDA)<br />

determine that it will not be profitable <strong>for</strong> seven years after FDA approval.<br />

It should be noted that incentives <strong>for</strong> developing orphan drugs are important, but only<br />

constitute a means to an end. 2 The ultimate success of such incentives should be<br />

measured in terms of the increase in life expectancy <strong>and</strong> quality of life of patients with<br />

rare diseases.<br />

Chapter 3 deals in more detail with the comparison between the policies of the FDA<br />

<strong>and</strong> EMEA.<br />

2.4.4 Marketing Authorisation<br />

Figure 2.1 presents data on the number of <strong>Orphan</strong> Designations <strong>and</strong> Marketing<br />

Authorisations <strong>for</strong> orphan drugs, issued by the FDA <strong>and</strong> the EMEA from 2001 until<br />

2007.<br />

Since the EU developed a Regulation in 2000 to promote research <strong>and</strong> development on<br />

orphan drugs, about 270 medicinal products received European <strong>Orphan</strong> Designation<br />

<strong>and</strong> 22 received Marketing Authorisation from EMEA by 2005. These numbers<br />

increased to 570 with <strong>Orphan</strong> Designation <strong>and</strong> 47 that received MA by December<br />

2008. In the USA, more than 240 orphan drugs reached the American market in the 20<br />

years following the <strong>Orphan</strong> Drug Act became law, <strong>and</strong> over 900 experimental orphan<br />

drugs are in the research pipeline. 19<br />

The rapid increase in the number of <strong>Orphan</strong> Drug Designations <strong>and</strong> Marketing<br />

Authorisations give rise to general concerns regarding the budget impact these drugs<br />

have <strong>and</strong> will have on the existing health care systems <strong>and</strong> health care payers (both<br />

public <strong>and</strong> private) <strong>and</strong> the extent to which the current governance support to these<br />

drugs is economically sustainable 2 14 18 .<br />

Figure 2.1 gives an overview of the number of approved <strong>Orphan</strong> Designations <strong>and</strong><br />

Marketing Authorisations by the FDA <strong>and</strong> the EMEA since 2001. The discrepancy<br />

between the numbers of FDA <strong>and</strong> EMEA is explained by the fact that the approvals in<br />

the USA started in 1984, increasing steadily over the years.<br />

d See inventories of 2001, 2002, 2005 17


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 13<br />

160<br />

140<br />

120<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

Figure 2.1 : <strong>Orphan</strong> Designation <strong>and</strong> Marketing Authorisation by the FDA<br />

<strong>and</strong> EMEA (2001-2007)<br />

2001 2002 2003 2004 2005 2006 2007<br />

2.4.5 Pricing<br />

OD: <strong>Orphan</strong> Designation;<br />

MA: Marketing Authorisation<br />

FDA - MA<br />

EMEA - MA<br />

FDA - OD<br />

EU - OD<br />

Prices of orphan drugs tend to be high. Both Genzyme <strong>and</strong> Shire, <strong>for</strong> example, are<br />

marketing some drugs in the EU with annual costs of € 200.000 to € 300.000 per<br />

patient (e.g. Aldurazyme® <strong>for</strong> mucopolysaccharidosis I <strong>and</strong> Fabrazyme® <strong>for</strong> Fabry<br />

disease). 20 There are several potential reasons <strong>for</strong> the high prices of orphan drugs. High<br />

prices may originate from marketing exclusivity, implying that no marketing<br />

authorization can be granted to a similar product with similar efficacy <strong>for</strong> the same<br />

therapeutic indication <strong>for</strong> a period of 10 years. The non-existence of an alternative<br />

treatment combined with the 10-year market exclusivity creates a monopoly <strong>for</strong> the<br />

company producing the orphan drug It should be noted, however, that marketing<br />

authorization can be granted to a clinically superior product, even if it is a similar<br />

product <strong>and</strong> market exclusivity can be reduced to 6 years if the criteria <strong>for</strong> orphan<br />

designation are no longer met. Also, the substantial costs of research <strong>and</strong> development<br />

have to be recouped from a small number of patients, thus resulting in high drug<br />

acquisition costs per patient. 13 However, orphan drugs do not always target a small<br />

number of patients.<br />

Certain products that were originally approved as orphan drugs, <strong>and</strong> as such benefited<br />

from special measures, later became top sellers either because the once rare condition<br />

they were intended to treat increased in frequency or because they proved also<br />

effective against more common disorderse.13 In these cases where profitability proves<br />

not to be a problem, public support is ex post deemed unjustified <strong>and</strong> critics in the US<br />

there<strong>for</strong>e urge <strong>for</strong> corrections to be made to the orphan drug legislation. In the EU,<br />

however, Article 8 of the Regulation provides <strong>for</strong> the possibility to reduce the<br />

marketing exclusivity to six years instead of ten years if, at the end of the fifth year, it is<br />

established that the product is sufficiently profitable. f Unsurprisingly, pharmaceutical<br />

companies lobby strongly against this article being put into practice <strong>and</strong> even argue that<br />

it should be eliminated completely. 21 Fact is also that there is no agreed definition of<br />

what is meant by “sufficiently profitable”.<br />

e Examples are Glivec® <strong>and</strong> Sutent®.<br />

f Situation on July 9th, 2009


14 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

2.4.6 Health technology assessment <strong>and</strong> reimbursement<br />

In a context of spiralling health care costs <strong>and</strong> limited resources, public policy makers<br />

<strong>and</strong> health care payers are increasingly using health technology assessments, including<br />

economic evaluation <strong>and</strong> budget impact analysis, to in<strong>for</strong>m reimbursement decisions.<br />

However, the use of health technology assessment in the field of orphan drugs <strong>for</strong><br />

reimbursement purposes is challenging <strong>for</strong> a number of reasons as described below.<br />

The reimbursement of orphan drugs is not regulated at EU level, but is a national<br />

responsibility of Member States. Once products have received Marketing Authorisation<br />

from EMEA, there are important differences to be noted among the EU Member States<br />

in terms of availability of these products on these markets, in the delays of availability<br />

between the Member States <strong>and</strong> in the prices of the same orphan medicine between the<br />

Member States. 19 Based on these observations, patient groups like the European<br />

Organisation <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong> (Eurordis) <strong>and</strong> the European Genetic Alliance Network<br />

(EGAN) are arguing <strong>for</strong> the elimination of regional <strong>and</strong> national differences in<br />

distribution, taxation <strong>and</strong> reimbursement policies, factors which combined explain the<br />

differences of up to 70% <strong>for</strong> the annual cost per patient of a given orphan medicine<br />

between various EU countries. 13<br />

Given their high price <strong>for</strong> an often modest health benefit orphan drugs are unlikely to<br />

be cost-effective, at least if the cost-effectiveness of an intervention is judged based on<br />

its cost per quality adjusted life year (QALY) gained in its neo-classical welfarist sense,<br />

<strong>and</strong> this cost-per-QALY is compared to a fixed threshold value. 22 If reimbursement<br />

decisions are primarily based on cost-effectiveness considerations <strong>and</strong> budget impact,<br />

orphan drugs will tend to fail these criteria. However, most often additional criteria that<br />

are not included in the traditional cost-per-QALY measure, are used to in<strong>for</strong>m<br />

reimbursement decisions. 22 For instance, the Pharmaceutical Benefits Advisory<br />

Committee of Australia also takes account of: the seriousness of the health condition;<br />

the availability of other therapies to treat the disease; <strong>and</strong> the cost to the patient if the<br />

drug is not reimbursed. 23 These criteria are particularly relevant to orphan drugs, which<br />

tend to target serious health conditions, make up the single strategy to treat a disease,<br />

<strong>and</strong> have a huge impact on patients’ health care expenditures if they would have to pay<br />

<strong>for</strong> the drugs themselves.<br />

With a view to assessing the effectiveness of an orphan drug, it is difficult to enrol a<br />

sufficient number of patients in clinical trials. As these diseases affect only few patients<br />

at a time, it is in many cases hardly possible to gather enough patients to achieve<br />

sufficient statistical power to demonstrate clinical effectiveness of a given treatment. 24<br />

Moreover, also because the disorders are rare, few medical centres will have sufficient<br />

long-term experience with affected patients to be able to describe the natural history of<br />

the diseases. Other authors also point out that in many rare disorders there is a lack of<br />

knowledge on disease processes, on the precise influence of genetics, on prevalence<br />

figures, <strong>and</strong> on how to conduct clinical trials. 19 These authors emphasise that increased<br />

ef<strong>for</strong>ts to address these issues are urgently needed at the European Community level.<br />

One of the authors there<strong>for</strong>e suggests to modify the review process <strong>for</strong> rare disease<br />

therapies: allow greater use of rational surrogate outcome measures if clinical efficacy<br />

data are incomplete, but require from industry to support a process of continuing<br />

review of clinical outcomes. A central component of the process would there<strong>for</strong>e be a<br />

commitment to ongoing evaluation of patients through registries designed to collect<br />

clinical in<strong>for</strong>mation on patients receiving the new therapy. 24<br />

A patient registry would allow regulatory authorities to follow up <strong>and</strong> evaluate the<br />

uncertainties surrounding longer-term effectiveness <strong>and</strong> cost-effectiveness of an orphan<br />

drug in the relevant population. 25 Such an approach would support the decision-making<br />

process <strong>and</strong> allow more timely access to orphan drugs <strong>for</strong> patients. Also, data on (cost-)<br />

effectiveness from patient registries can be used by researchers <strong>and</strong> clinicians to in<strong>for</strong>m<br />

clinical practice <strong>and</strong> prescribing guidelines. Such patient registries could even be<br />

integrated with national pharmacovigilance systems providing in<strong>for</strong>mation about adverse<br />

events associated with orphan drugs. However, it should be noted that there are<br />

challenges involved in setting up a patient registry <strong>and</strong> analysing registry data. They<br />

could also be different if set-up independently or by industry.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 15<br />

The patient registry may be biased as the patient aetiology <strong>and</strong> disease severity change<br />

over time. Also, as patient registries collect data on an orphan drug, but not on<br />

alternative treatments, they only provide partial in<strong>for</strong>mation to calculate the<br />

incremental cost-effectiveness of the orphan drug relative to an alternative treatment.<br />

Furthermore, new treatment strategies may become available during the period<br />

covered by the registry. There<strong>for</strong>e, patient registries need to be set up <strong>and</strong> developed<br />

in a flexible way to be able to account <strong>for</strong> changes in patient population <strong>and</strong> treatment<br />

strategies over their lifetime.<br />

For instance, the MPSI Registry is an ongoing, observational database that tracks natural<br />

history <strong>and</strong> outcomes of patients with MPSI g . Initiated worldwide in April 2003, data<br />

from over 718 patients with MPSI have been collected from physicians in over 30<br />

countries as of May 2008.<br />

Reimbursement may not only depend on the value <strong>for</strong> money of an orphan drug at the<br />

time of the reimbursement application, but also on its value after a number of years<br />

following the admission to the reimbursement system. Under such a system of<br />

conditional reimbursement, pharmaceutical companies need to explore setting up<br />

patient registries to in<strong>for</strong>m the post-launch cost-effectiveness of an orphan drug.<br />

Reimbursement authorities may also wish to consider a risk-sharing scheme between<br />

drug sponsor <strong>and</strong> government based on a registry system whereby survival outcomes<br />

are linked to future drug prices. 25 Risk-sharing agreements allow authorities to balance<br />

the uncertainty of long-term cost-effectiveness with the need to provide equitable<br />

access to potentially effective but expensive orphan drugs. Such agreements may incite<br />

the drug sponsor to promote responsible prescribing of orphan drugs; provide a<br />

guarantee on health outcomes with a view to attaining predictable health gains <strong>for</strong> a<br />

given drug expenditure; <strong>and</strong> share the budgetary risks between authorities <strong>and</strong> the drug<br />

sponsor. However, risk-sharing agreements entail that structures are set in place that<br />

safeguard the objective of computing the post-marketing cost-effectiveness of orphan<br />

drugs based on a representative <strong>and</strong> unbiased sample. Also, risk-sharing agreements<br />

should be flexible to reflect the introduction of new treatment strategies over the<br />

monitoring period of the agreement. Nevertheless, Owen et al. conclude that such<br />

schemes may allow to balance the uncertainty of long-term cost-effectiveness<br />

with the public dem<strong>and</strong> <strong>for</strong> equitable <strong>and</strong> timely access to new orphan drugs,<br />

on the condition that some issues like governance, privacy, ethics review, timely <strong>and</strong><br />

accurate data, related to such registry system are adequately addressed. 25<br />

2.5 CONCLUSIONS<br />

A wide variety of definitions of rare diseases <strong>and</strong> of orphan drugs are used in the<br />

legislation of various countries. Also, a number of challenges exist with respect to the<br />

development, Marketing Authorisation, pricing, reimbursement <strong>and</strong> post-marketing<br />

follow-up of orphan drugs. As a result, the need is expressed <strong>for</strong> more transnational<br />

cooperation <strong>and</strong> the building of an active <strong>and</strong> international community able to act <strong>and</strong><br />

address the economic <strong>and</strong> intellectual ef<strong>for</strong>ts towards solving the most pressing<br />

difficulties as described above. 19<br />

g http://www.lsdregistry.net/mpsiregistry/


16 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Key points<br />

• Different definitions of rare diseases <strong>and</strong> of orphan drugs are used in the<br />

existing legislation of various countries <strong>and</strong> in literature. For the purpose of<br />

the present study, EU official definitions <strong>for</strong> rare diseases <strong>and</strong> <strong>for</strong> orphan<br />

drugs are adopted.<br />

• <strong>Rare</strong> diseases are life-threatening or chronically debilitating conditions with<br />

a prevalence of 50 out of 100 000 or less.<br />

• Due to their relatively low prevalence, rare diseases have traditionally been<br />

neglected by large parts of the scientific, medical <strong>and</strong> political communities.<br />

As a result, patients suffering from rare diseases may experience unequal<br />

access to treatment in comparison to patients suffering from ‘common’<br />

diseases.<br />

• In the EU, orphan drugs are considered differently from other drugs <strong>for</strong><br />

reasons of absence of economical viability under normal market conditions<br />

<strong>and</strong> because of considerations of patient equity. With a view to supporting<br />

the development of orphan drugs, the EU has put in place a number of<br />

incentives.<br />

• However, the resulting rapid increase in the number of orphan drugs<br />

obtaining designations has given rise to concerns about the overall potential<br />

budget impacts on health care systems <strong>and</strong> health care payers.<br />

• Reimbursement decisions <strong>for</strong> pharmaceutical products are also based on<br />

cost-effectiveness considerations, but orphan drugs will tend to fail these<br />

criteria because the cost of orphan drug treatments is usually high <strong>for</strong> the<br />

benefits they offer compared to many non-rare disease treatments.<br />

Additional criteria, such as the seriousness of the disease <strong>and</strong> the availability<br />

of other therapies, can become more important in reimbursement decisions<br />

of orphan drugs but are generally not incorporated in a st<strong>and</strong>ard economic<br />

evaluation.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 17<br />

3 POLICY DESCRIPTION<br />

3.1 INTRODUCTION<br />

The process from <strong>Orphan</strong> Designation to Marketing Authorisation is governed by the<br />

European Medicines Agency (EMEA) in Europe <strong>and</strong> the Food <strong>and</strong> Drug Administration<br />

(FDA) in the United States. This chapter compares the EMEA <strong>and</strong> FDA procedures with<br />

a view to identifying differences <strong>and</strong> discussing the implications of different approaches<br />

to <strong>Orphan</strong> Designation <strong>and</strong> Marketing Authorisation.<br />

3.2 EMEA PROCESS: FROM ORPHAN DESIGNATION TO<br />

MARKETING AUTHORISATION<br />

3.2.1 Presentation of EMEA<br />

“The European Medicines Agency is a decentralised body of the European Union with<br />

headquarters in London. Its main responsibility is the protection <strong>and</strong> promotion of public <strong>and</strong><br />

animal health, through the evaluation <strong>and</strong> supervision of medicines <strong>for</strong> human <strong>and</strong> veterinary<br />

use”. 26<br />

Until recently, the possibility also existed to obtain a Marketing Authorisation by a<br />

mutual recognition procedure. Since 2005, only the central procedure at EMEA can be<br />

used to register a product <strong>and</strong> obtain Marketing Authorisation.<br />

The two main actors in the orphan drug procedures are the Committee <strong>for</strong> <strong>Orphan</strong><br />

Medicinal Products (COMP) <strong>and</strong> the Committee <strong>for</strong> Human Medicinal Products<br />

(CHMP).<br />

The COMP is EMEA’s committee responsible <strong>for</strong> examining the applications <strong>for</strong> <strong>Orphan</strong><br />

Designation: the European Commission (EC) will approve or reject an application based<br />

on the COMP’s opinion. The COMP has two main activities:<br />

Scientific evaluation Public Health Activities<br />

• To examine applications <strong>for</strong> <strong>Orphan</strong><br />

Drug Designations<br />

• Protocol assistance<br />

• Re-evaluation of significant benefit<br />

during Marketing Authorisation<br />

registration<br />

• Post-Marketing Authorisation review<br />

every 5 years<br />

• Advise the EC on the establishment <strong>and</strong><br />

development of a policy on orphan<br />

medicinal products <strong>for</strong> the EU<br />

• Assist the EC in liaising internationally on<br />

matters relating to orphan medicinal<br />

products, <strong>and</strong> in liaising with patient support<br />

groups<br />

• Assist the EC in drawing up detailed<br />

guidelines<br />

• EU expert network <strong>and</strong> visibility<br />

The COMP is composed of:<br />

• one chairperson <strong>and</strong> one vice-chairperson;<br />

• one member per Member State (27 Member States in 2008);<br />

• three members representing patient organisations (nominated by the EC);<br />

• three members recommended by EMEA (nominated by the EC);<br />

• one non-voting member per EEA-EFTA h state (Norway <strong>and</strong> Icel<strong>and</strong> in<br />

2008).<br />

The members are appointed by their country, while the chairperson <strong>and</strong> vicechairperson<br />

are elected by <strong>and</strong> from the COMP members on basis of a brief resume of<br />

the c<strong>and</strong>idates <strong>and</strong> with an absolute majority i .<br />

h<br />

EEA-EFTA: European Economic Area – European Free Trade Association<br />

i<br />

An absolute majority is obtained when more than 50% of the members has voted in favour of the<br />

c<strong>and</strong>idate.


18 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

All the members are appointed <strong>for</strong> a period of three years with possibility of renewal<br />

(the same applies <strong>for</strong> the CHMP members).<br />

The CHMP is the committee responsible <strong>for</strong> examining the applications <strong>for</strong> Marketing<br />

Authorisation <strong>for</strong> all medicinal products, but also <strong>for</strong> the post-authorisation follow-up.<br />

The CHMP is composed of:<br />

• a chairman, elected by <strong>and</strong> from the CHMP members;<br />

• one member (<strong>and</strong> an alternate) per Member State (27 Member States in<br />

2008);<br />

• one member (<strong>and</strong> an alternate) per EEA-EFTA state (Norway <strong>and</strong> Icel<strong>and</strong><br />

in 2008);<br />

• up to five co-opted members (experts recruited to gain additional<br />

expertise in a particular scientific area).<br />

The European <strong>Orphan</strong> Drug policy has been regulated by Regulation (EC) No 141/2000 5<br />

on orphan medicinal products. There are two steps to be taken be<strong>for</strong>e an orphan drug<br />

is admitted on the market:<br />

• Step 1: <strong>Orphan</strong> Designation: a medicinal product receives the orphan<br />

status – linked to incentives.<br />

• Step 2: Marketing Authorisation: is the marketing approval of a drug <strong>for</strong><br />

an orphan condition. It becomes orphan drug <strong>and</strong> receives market<br />

exclusivity.<br />

3.2.2 Applying <strong>for</strong> <strong>Orphan</strong> Designation<br />

3.2.2.1 Conditions to be fulfilled<br />

A medicinal product can obtain the designation of orphan medicinal product if it<br />

provides treatment <strong>for</strong> a rare disease. In order <strong>for</strong> a pharmaceutical firm to be able to<br />

apply <strong>for</strong> an <strong>Orphan</strong> Designation, two conditions must be fulfilled (see art. 3 of<br />

Regulation (EC) no 141/2000):<br />

• the medicinal product is intended <strong>for</strong> the diagnosis, prevention or<br />

treatment of a life-threatening or chronically debilitating condition that<br />

either affects less than 5 in 10,000 persons of the Community; or that<br />

without incentives it is unlikely that the marketing of the medicinal<br />

product would generate sufficient return to justify the expenditure;<br />

• a satisfactory method <strong>for</strong> diagnosis, prevention or treatment of the<br />

condition does not exist.<br />

The COMP will essentially take decisions at two levels corresponding to the two<br />

conditions defined in the above-mentioned article of the Regulation:<br />

Level 1: Prevalence or Insufficient return<br />

Level 2: Absence of solution or Significant benefit<br />

Other criteria to obtain <strong>Orphan</strong> Designation are: the “medical plausibility” of the<br />

condition <strong>and</strong> sub-setting.<br />

In case of insufficient return it is unlikely that the expected return would justify the<br />

required investment <strong>and</strong> so it is unlikely that the sponsor would be prepared to make<br />

the investment. 27<br />

Absence of solution means that there is no alternative treatment available, while<br />

significant benefit 28 means that the drug has a clinically relevant advantage or major<br />

contribution to patient care compared to existing satisfactory methods <strong>for</strong> diagnosis,<br />

prevention or treatment. The significant benefit can be related to:<br />

• improved efficacy <strong>and</strong> / or safety;<br />

• ease of self administration: leading to a major contributions to patient<br />

care;


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 19<br />

• new mechanism of action: of which the efficacy will have to be<br />

demonstrated. This new mechanism opens possibilities <strong>for</strong> drug<br />

combination <strong>and</strong> can be designated as a therapeutic alternative 29 ;<br />

• reduced availability of the base materials;<br />

• reduced availability of the product in the EU Member States (not possible<br />

after centralised registration).<br />

Significant benefit must be confirmed by the sponsors during registration application,<br />

<strong>and</strong> questions about it must have been answered during protocol assistance. Appraisal<br />

of the criterion thus occurs at three times: 1. first application at COMP <strong>for</strong> <strong>Orphan</strong><br />

Designation; 2. protocol assistance prior to Marketing Authorisation; 3. MA registration<br />

application (compulsory). The COMP will assess if the significant benefit can be<br />

confirmed by available data <strong>and</strong>/or evidence supplied by the applicant (at the moment<br />

the CHMP takes its decision).<br />

A subset 28 is a separated part of a (frequently occurring) disease, having an own<br />

pharmacotherapeutic treatment <strong>and</strong> without this subset the drug would have no effect<br />

in the remaining population. Sub-setting is rarely accepted. Are not accepted:<br />

• different levels of seriousness or localisation of a disease;<br />

• the subset is based on a (post-hoc) analysis of the study of a product that<br />

should function <strong>for</strong> the whole group.<br />

Sub-setting can lead to so-called “salami-slicing”: this is creating artificial subsets of a<br />

non-orphan condition, by basing the prevalence criterion on an unreal subpopulation.<br />

The aim is to obtain market exclusivity, a decrease of the costs <strong>and</strong> obligations linked to<br />

the registration dem<strong>and</strong>, <strong>and</strong> an increase of the exclusivity through new subpopulations<br />

(also known as the “evergreening tactic”).<br />

Medically plausible subsets are based on the real disease process; the seriousness of<br />

the condition; the characteristics of the drug; the working mechanism <strong>and</strong> the unique<br />

characteristics of the patient population. In order <strong>for</strong> a subset to be accepted, the<br />

sponsor must justify the medical plausibility why the drug should be restricted to the<br />

sub-set. 30<br />

In practice, of the 541 molecules or products that received the designation, 540<br />

obtained it based on the criterion “prevalence”, <strong>and</strong> one product obtained designation<br />

based on the criterion of “return on investment” j (a second application was withdrawn<br />

by the sponsor 30 ).<br />

3.2.2.2 <strong>Orphan</strong> Designation Procedure<br />

The application dossier must include:<br />

• the name or corporate name <strong>and</strong> permanent address of the sponsor;<br />

• active ingredients of the medicinal product;<br />

• proposed therapeutic indication;<br />

• a justification that the two conditions mentioned in article 3 are met;<br />

• <strong>and</strong> a description of the stage of development, including the therapeutic<br />

indications expected.<br />

The <strong>Orphan</strong> Designation procedure is described in figure 3.1. This is the centralised<br />

procedure which is compulsory <strong>for</strong> orphan drugs since 2005. k<br />

The COMP will adopt its opinion with a consensus, <strong>and</strong> if impossible with a two-thirds<br />

majority. The European Commission may take a draft decision that is different of the<br />

COMP’s opinion, but this decision must be approved by the Council of the EU (See art.<br />

73 of Regulation (EC) 2309/93) 31 .<br />

j This orphan drug designation was obtained <strong>for</strong> the treatment of neglected disease (as opposed to rare<br />

disease).<br />

k For non-orphan drugs, it is also possible to proceed through the mutual recognition procedure <strong>and</strong> the<br />

decentralised or national procedure.


20 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

90 days<br />

30 days<br />

Following the publication of the orphan product in the Community Register of <strong>Orphan</strong><br />

Medicinal Products, the sponsor will annually submit to the EMEA a report on the state<br />

of development of the product (See art. 5§10 of Regulation (EC) 141/2000) 5 .<br />

Figure 3.1 : <strong>Orphan</strong> Designation Procedure<br />

Sponsor submits application <strong>for</strong>m to the EMEA<br />

anytime during the development process of the<br />

product<br />

- Appointment of 2 coordinators:<br />

COMP member + EMEA staff member<br />

- EMEA verifies validity<br />

- The EMEA coordinator prepares a<br />

summary report of the application <strong>for</strong><br />

the COMP members start of the<br />

evaluation time-table<br />

COMP must give an opinion within 90 days of<br />

reception based on the summary report <strong>and</strong> its<br />

members’ comments<br />

European Commission takes a decision based on<br />

the opinion within 30 days <strong>and</strong> in<strong>for</strong>ms sponsor,<br />

EMEA <strong>and</strong> Member States<br />

The designated orphan medicinal product is<br />

registered in the Community Register of <strong>Orphan</strong><br />

Medicinal Products<br />

Post-designation: the sponsor submits an annual<br />

report on the state of development of the<br />

product (this is an administrative procedure – no<br />

sanction)<br />

Pre-submission meeting<br />

is possible<br />

Sponsor may appeal<br />

within 90 days following<br />

the opinion<br />

In order to encourage research <strong>and</strong> development of orphan medicinal products, the<br />

<strong>Orphan</strong> Regulation incorporates five incentives. The first incentive can take place<br />

be<strong>for</strong>e or after applying <strong>for</strong> <strong>Orphan</strong> Designation: thanks to the protocol assistance (1),<br />

the company may request scientific advice of the EMEA on the conduct of various tests<br />

<strong>and</strong> trials necessary to demonstrate the quality, safety <strong>and</strong> efficacy of the medicinal<br />

product. Once the medicinal product has been given the status of orphan, it has direct<br />

access to the centralised procedure (2) <strong>for</strong> the application <strong>for</strong> Marketing Authorisation.<br />

If this latter is granted, the orphan medicinal product receives a 10-year market<br />

exclusivity (3) meaning that similar products have no access to the market (unless they<br />

have a significant benefit or are superior).<br />

<strong>Orphan</strong> medicinal products will also benefit from fee reductions (4) <strong>for</strong> centralised<br />

applications <strong>and</strong> obtain grants within the framework of EU-funded research (5) as well<br />

as priority access to EU research programs.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 21<br />

The market exclusivity can be brought back to six years if the sponsor having applied<br />

first <strong>for</strong> a designation gives its consent that a second sponsor can obtain the same<br />

designation; there is a lack of drug supply; or if the new drug is safer, more effective of<br />

clinically superior. 29 The market exclusivity may also be withdrawn after six years if the<br />

medicinal product no longer meets the two conditions necessary to obtain <strong>Orphan</strong><br />

Drug Designation.<br />

In 2007, 4.89 million € of fee reductions was granted through the fee reduction<br />

mechanisms (applicable after <strong>Orphan</strong> Drug Designation is obtained).<br />

Figure 3.2 : Overview of incentives <strong>and</strong> other compensations<br />

Incentives:<br />

1. 10 years market exclusivity<br />

2. Protocol <strong>and</strong> scientific assistance<br />

3. Financial incentives on a national basis<br />

4. Direct access to centralised procedure<br />

5. Access to EU-funded research<br />

Other compensations:<br />

1. The fee reductions can be the following: 16<br />

o 100% reduction <strong>for</strong> protocol assistance <strong>and</strong> follow-up;<br />

o 100% reduction <strong>for</strong> pre-authorisation inspections;<br />

o 50% reduction <strong>for</strong> applications <strong>for</strong> Marketing Authorisation;<br />

o 50% reduction <strong>for</strong> post-authorisation activities, including annual fees,<br />

in the first year after granting of a Marketing Authorisation.<br />

2. 12-year market exclusivity if paediatric orphan drug (Paediatric development<br />

since 1/7/2008)<br />

3. Guidance <strong>for</strong> clinical trials in small populations 32 in order to increase the<br />

efficiency of the design <strong>and</strong> the analysis<br />

Figure 3.3 : Use of EU special funding contribution <strong>for</strong> orphan medicines<br />

(2007)<br />

68%<br />

5% 3%<br />

24%<br />

Marketing Authorisation<br />

Protocol assistance<br />

Inspections<br />

Post-authorisation<br />

Source: EMEA. Annual report of the European Medicines Agency 2007. Doc. ref.:<br />

EMEA/MB/17464/2008 13 May 2008. Available from<br />

[Last accessed:<br />

10/12/2008].


22 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

The table in figure 3.4 gives an overview of the yearly number of applications <strong>for</strong><br />

<strong>Orphan</strong> Designation since 2000. Of 896 initial applications 226 have been withdrawn.<br />

92% of the remaining applications have received a positive COMP opinion <strong>and</strong> 89% a<br />

final designation.<br />

Figure 3.4 : <strong>Orphan</strong> Designation between 2000 <strong>and</strong> November 2008<br />

Year Applications<br />

submitted<br />

Positieve<br />

COMP<br />

Opinions<br />

Applications<br />

withdrawn<br />

Final negative<br />

COMP Opinions<br />

Designations<br />

granted by<br />

the<br />

Commission<br />

2008 119 86 31 1 73<br />

2007 125 97 19 1 98<br />

2006 104 81 20 2 80<br />

2005 118 88 30 0 88<br />

2004 108 75 22 4 72<br />

2003 87 54 41 1 55<br />

2002 80 43 30 3 49<br />

2001 83 64 27 1 64<br />

2000 72 26 6 0 14<br />

Total 896 614 226 13 593<br />

Source: Committee <strong>for</strong> <strong>Orphan</strong> Medicinal Products. January 2009 Plenary Meeting, Monthly<br />

Report. EMEA. Doc. Ref.: EMEA/COMP/694107/2008. 7 January 2009. Available from<br />

[Last accessed: 10/3/2009].<br />

3.2.3 Applying <strong>for</strong> Marketing Authorisation<br />

Be<strong>for</strong>e applying <strong>for</strong> a Marketing Authorisation, the pharmaceutical company can request<br />

scientific advice <strong>and</strong> protocol assistance from the Scientific Advice Working Party<br />

(SAWP), part of the CHMP. The advice is given <strong>for</strong> the conduct of tests <strong>and</strong> trials in<br />

order to demonstrate the quality, safety <strong>and</strong> efficacy of the medicinal product (Art. 6<br />

Regulation (EC) No. 141/2000) 5 , while the assistance provides guidance <strong>and</strong> verifies the<br />

criteria.<br />

Figure 3.5 : Scientific-advice <strong>and</strong> protocol-assistance requests received, 2005-2007<br />

2007<br />

2006<br />

2005<br />

58<br />

58<br />

68<br />

0 50 100 150 200 250<br />

Scientific-advice <strong>and</strong> follow-up requests Protocol-assistance <strong>and</strong> follow-up requests<br />

Source: EMEA. Annual report of the European Medicines Agency 2007. Doc. ref.:<br />

EMEA/MB/17464/2008 13 May 2008. Available from<br />

[Last accessed:<br />

10/12/2008].<br />

136<br />

201<br />

213


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 23<br />

l CHMP/EWP/83561/2005<br />

The SAWP’s tasks are to: 33<br />

• provide the CHMP an integrated view about the quality, pre-clinical <strong>and</strong><br />

clinical safety including pharmacovigilance <strong>and</strong> risk/minimisation aspects,<br />

<strong>and</strong> efficacy, relating to the development of orphan medicinal products;<br />

• provide protocol assistance to the CHMP as regards the demonstration of<br />

significant benefit relating to orphan medicinal products;<br />

• provide advice on applying <strong>for</strong> a conditional MA or MA under exceptional<br />

circumstances;<br />

• provide advice on the design of trials to assess safety <strong>and</strong> efficacy in a new<br />

indication expected to bring significant clinical benefit compared to<br />

existing therapies;<br />

• pay special attention to development <strong>and</strong> methodology issues of products<br />

intended <strong>for</strong> small populations.<br />

In 2006, the CHMP developed guidelines on clinical trials in small populations. l They<br />

came into effect on February 1st, 2007. The guidelines acknowledge that in<br />

circumstances where only few patients are affected by a disease, a trial enrolling several<br />

hundred patients may not be practical or possible. Meanwhile it is stated that “most<br />

orphan drugs <strong>and</strong> paediatric indications submitted <strong>for</strong> regulatory approval are based on<br />

r<strong>and</strong>omised controlled trials that follow generally accepted rules <strong>and</strong> guidance.” The<br />

guidelines state that “deviation from such st<strong>and</strong>ards is, there<strong>for</strong>e, uncommon <strong>and</strong><br />

should only be considered when completely unavoidable <strong>and</strong> would need to be<br />

justified.”<br />

After having received the <strong>Orphan</strong> Designation the sponsor can apply <strong>for</strong> a Marketing<br />

Authorisation (the procedure is presented in figure 3.6). This can only be done through<br />

the centralised procedure at the EMEA. For normal drugs, a national procedure exists<br />

as well. Within 210 days the CHMP gives a final opinion that is transmitted to the<br />

European Commission who will take the concluding decision.<br />

The MA procedure consists of three steps:<br />

1. Pre-submission<br />

2. Primary evaluation<br />

3. Secondary evaluation<br />

An accelerated review of the medicinal product is possible when decided by the CHMP.<br />

The product must fulfil three conditions:<br />

• the condition is life threatening or serious;<br />

• there is no effective therapeutic alternative;<br />

• <strong>and</strong> the drug is expected to have a high therapeutic benefit. 34<br />

1. Pre-submission phase<br />

The sponsor (named hereafter ‘applicant’) sends a letter of intent to the CHMP<br />

together with a fee <strong>for</strong> the examination. The CHMP examines the validity of the<br />

application:<br />

• examination of the submitted particulars <strong>and</strong> documents;<br />

• it may request that the medicinal product be tested;<br />

• <strong>and</strong> it may request that the applicant supplements the particulars<br />

accompanying the application within a specific time period.<br />

A rapporteur <strong>and</strong> a co-rapporteur will be appointed to evaluate the MA application. The<br />

appointment is based on the best available expertise. Each rapporteur will have an<br />

assessment team composed of assessors of the national authorities <strong>and</strong> can appoint<br />

experts if necessary.


24 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Within EMEA, a product team leader <strong>and</strong> his/her team are appointed in order to<br />

prepare the documents of the CHMP <strong>and</strong> to liaise between the applicant <strong>and</strong> the<br />

CHMP.<br />

2. Primary evaluation<br />

On Day 80 of the MA process, the two rapporteurs each produce an assessment report<br />

in which is given a detailed overview of the quality, clinical <strong>and</strong> non-clinical data given in<br />

the submission file, <strong>and</strong> possibly a proposal of list of questions.<br />

The assessment reports are sent to the CHMP members <strong>for</strong> comments <strong>and</strong> to the<br />

applicant <strong>for</strong> in<strong>for</strong>mation. The reports are also peer reviewed by a CHMP member <strong>and</strong><br />

by the EMEA product team leader in order to see if they are consistent <strong>and</strong> if there is a<br />

sufficient level of detail.<br />

A list of questions is produced by the CHMP <strong>and</strong> sent to the applicant on Day 120.<br />

3. Secondary evaluation<br />

A Joint Assessment Report is produced by the rapporteurs following the reception of<br />

the responses of the applicant. The report is sent to the CHMP on Day 150 <strong>for</strong><br />

comments resulting in a list of outst<strong>and</strong>ing issues to be sent to the applicant on Day<br />

180. Following the reception of the applicant’s responses, a second joint assessment<br />

report is sent to the CHMP members.<br />

The different assessment reports <strong>and</strong> the lists of questions serve as a basis <strong>for</strong> the<br />

redaction of the CHMP assessment report which underpins the CHMP opinion. The<br />

CHMP opinion is based on the examination of the risk-benefit balance of the medicinal<br />

product; this is an evaluation of the positive therapeutic effects of the medicinal product<br />

in relation to the following risks:<br />

• any risk related to the quality, safety or efficacy of the medicinal product<br />

as regards patients' health or public health;<br />

• any risk of undesirable effects on the environment.<br />

The CHMP issues a positive or negative opinion based on the risk-benefit ratio. There is<br />

no comparison with other drugs.<br />

The opinion is sent to the European Commission who takes the final decision. Refusal<br />

reasons are either that the quality, safety or efficacy of the product is not<br />

demonstrated; or that particulars or documents are incorrect. If the EC decision is<br />

different from the CHMP’s opinion, then it will be sent to the Member States. The<br />

applicant is notified in both cases.<br />

The CHMP assessment report also serves as a basis <strong>for</strong> the redaction of the European<br />

Public Assessment Report (EPAR) which is published on EMEA’s website <strong>and</strong> available<br />

<strong>for</strong> the public.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 25<br />

MARKETING AUTHORISATION<br />

REDACTION EPAR<br />

Figure 3.6 : Marketing Authorisation Procedure<br />

Day 1 Start procedure<br />

Day 80 Assessment report of rapporteur <strong>and</strong> corapporteur<br />

sent to CHMP<br />

Day 120 List of questions of CHMP sent to applicant<br />

Day 150 Joint assessment report from both<br />

rapporteurs based on applicants’ answers<br />

Day 180 List of outst<strong>and</strong>ing issues<br />

Day 181 Second joint report of rapporteurs<br />

Day 210 Adoption of CHMP assessment report <strong>and</strong><br />

opinion<br />

To applicant: is there any in<strong>for</strong>mation that should be<br />

deleted because commercial sensitive?<br />

Agreement on content<br />

EPAR DRAFT I (assessment report <strong>and</strong> EPAR Summary<br />

(positive opinion) / Q&A (negative opinion)<br />

Comments CHMP members<br />

Comments CHMP members<br />

If comments from members CHMP <br />

EPAR DRAFT II<br />

Adoption EPAR DRAFT I/II by CHMP Finalisation<br />

EPAR<br />

European Commission’s<br />

decision


26 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

The MA procedure is the same <strong>for</strong> all human medicinal products, orphan <strong>and</strong> nonorphan<br />

drugs: EMEA evaluates the quality, safety <strong>and</strong> efficacy of the drug. The only<br />

differences <strong>for</strong> orphan drugs are:<br />

1. the involvement of the COMP who will review the significant benefit<br />

criterion: are the criteria on which the decision <strong>for</strong> the <strong>Orphan</strong> Designation<br />

were taken still valid? This takes place when the CHMP prepares its opinion.<br />

2. the existence of guidelines <strong>for</strong> clinical trials in small populations, which are<br />

used as a basis to assess the clinical evidence provided.<br />

The MA is granted if:<br />

1. The medicinal product contains a new active substance which was not<br />

authorised in the Community; or<br />

2. The applicant shows that the medicinal product constitutes a significant<br />

therapeutic, scientific or technical innovation or that the granting of<br />

authorisation is in the interests of patients at Community Level.<br />

A medicinal product cannot receive a MA <strong>for</strong> an orphan indication <strong>and</strong> <strong>for</strong> a nonorphan<br />

indication. In case of conflict, the orphan indication will have to be disposed of<br />

or the medicinal product will have to request MA under a different name. For example,<br />

the drugs Viagra® <strong>and</strong> Revatio® have the same composition, but the first is reimbursed<br />

<strong>for</strong> a non-orphan indication, the second <strong>for</strong> an orphan indication (Pulmonary Arterial<br />

Hypertension).<br />

The MA is valid <strong>for</strong> five years <strong>and</strong> can be renewed <strong>for</strong> five-year periods after review<br />

(Art. 13 Regulation (EC) No. 2309/93). 31<br />

There is a possible access to accelerated review to MA if duly substantiated by the<br />

sponsor (150 days instead of 210 days). The review can be requested <strong>for</strong> medicinal<br />

products <strong>for</strong> human use which are of major interest from the point of view of public<br />

health <strong>and</strong> of therapeutic innovation.<br />

In 2008, fourteen MA applications have been submitted <strong>for</strong> orphan medicinal products.<br />

Seven have received a positive opinion <strong>and</strong> two a negative opinion. Six sponsors have<br />

withdrawn their application prior to the opinion. The European Commission has<br />

granted five MA. 35<br />

Figure 3.7 : Overview of Marketing Authorisations <strong>for</strong> <strong>Orphan</strong> <strong>Drugs</strong> (2001-<br />

2008)<br />

Number of approved MA per year<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

2001 2002 2003 2004 2005 2006 2007 2008<br />

Number of approved MA per year Total number of MA<br />

Source: DG Enterprise EC. Register of designated <strong>Orphan</strong> Medicinal Products.<br />

. 11/3/2009.<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

0<br />

Total number of MA per year


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 27<br />

Figure 3.8 : Total of <strong>Orphan</strong> <strong>Drugs</strong> per therapeutic area (December 2008)<br />

Cardiovascular<br />

& respiratory<br />

14%<br />

Nervous<br />

system<br />

8%<br />

Haematology<br />

16%<br />

Other<br />

6%<br />

Oncology<br />

29%<br />

Endocrinology /<br />

metabolism<br />

27%<br />

Source: EMEA. List of orphan-designated authorised medicines 6/11/2008. Available from<br />

[Last accessed: 7/5/2009].<br />

There are three types of MA: the normal MA, the conditional MA <strong>and</strong> the MA under<br />

exceptional circumstances.<br />

The normal MA is valid <strong>for</strong> five years <strong>and</strong> may be renewed on the basis of a reevaluation<br />

by EMEA of the risk-benefit balance. After this renewal, the MA will be valid<br />

<strong>for</strong> an unlimited period of time unless decided otherwise. The medicinal product must<br />

be placed on the market within three years otherwise the Authorisation is no longer<br />

valid. The same applies when the product has been unavailable <strong>for</strong> three consecutive<br />

years.<br />

The conditional MA is regulated by Regulation (EC) 726/2004. This second type of<br />

MA is granted on the basis of less complete clinical data. Conditions are that the riskbenefit<br />

balance is positive, there is a benefit to public health of immediate market<br />

availability (outweighing the risks inherent to the fact that additional data are still<br />

required) <strong>and</strong> that unmet medical needs will be fulfilled. The conditional MA is valid <strong>for</strong><br />

one year <strong>and</strong> can be renewed. Once the missing data have been completed, the drug<br />

will receive a normal MA.<br />

The MA under exceptional circumstances is given when comprehensive data<br />

cannot be provided because of the small study population. This MA will be reviewed<br />

annually to reassess the risk-benefit balance based on follow-up studies including<br />

pharmacovigilance studies. The orphan drugs Tracleer® <strong>and</strong> Fabrazyme® have received<br />

normal MA after fulfilling the data.<br />

As of December 2008, there was one orphan drug with a conditional MA <strong>and</strong> sixteen<br />

with an exceptional MA. This means that five out of eight orphan drugs are authorised<br />

under exceptional circumstances. Following figure shows that the exceptional status is<br />

given at least once yearly <strong>and</strong> is not something which was mostly used in the early years<br />

of the legislation.


28 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Figure 3.9: Overview of Exceptional Marketing Authorisations (2001-2008)<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

2001 2002 2003 2004 2005 2006 2007 2008<br />

Source: DG Enterprise EC. Register of designated <strong>Orphan</strong> Medicinal Products.<br />

. 11/3/2009.<br />

3.2.4 Compassionate use<br />

Compassionate use is possible <strong>for</strong> new medicinal products to be approved through the<br />

centralised procedure of EMEA: there<strong>for</strong>e three conditions states in art. 83 of<br />

Regulation (EC) No 726/2004 36 must be fulfilled:<br />

• The medicinal product is to be made available to “patients with a<br />

chronically or seriously debilitating disease, or a life threatening disease,<br />

<strong>and</strong> who cannot be treated satisfactorily by an authorised medicinal<br />

product” in the EU,<br />

• The compassionate use programme is intended <strong>for</strong> a “group of patients”,<br />

• The medicinal product is either “the subject of an application <strong>for</strong> a<br />

centralised Marketing Authorisation in accordance with Article 6 of<br />

Regulation (EC) No 726/2004 or is undergoing clinical trials” in the EU or<br />

elsewhere.<br />

The aim of the compassionate use programme is to facilitate access <strong>for</strong> patients to a<br />

new medicinal product.<br />

There are also compassionate use programmes at a national level which differ between<br />

Member States. Some of them are addressed in the next chapter.<br />

3.3 FDA PROCESS: FROM ORPHAN DESIGNATION TO<br />

MARKETING AUTHORISATION<br />

3.3.1 Presentation of the FDA<br />

The <strong>Orphan</strong> Drug Act signed in 1983 was the first orphan drug legislation adopted in<br />

the world. It defines an orphan drug as a drug that is intended to treat a condition<br />

affecting fewer than 200,000 persons in the United States, or which will not be<br />

profitable within 7 years following approval by the U.S. Food & Drug Administration<br />

(FDA). 37<br />

The legislation regulating the <strong>Orphan</strong> <strong>Drugs</strong> Policy in the United States can be found in<br />

the Code of Federal Regulations (CFR), Title 21, Part 316: <strong>Orphan</strong> <strong>Drugs</strong>. 38 The<br />

authority in charge is the Office of <strong>Orphan</strong> Products Development (OOPD) of the<br />

FDA. The OOPD’s primary objective is to promote the development of products that<br />

demonstrate promise of the diagnosis <strong>and</strong>/or treatment of rare diseases or conditions. 39<br />

In order to obtain marketing approval, a drug first has to obtain <strong>Orphan</strong> Drug<br />

Designation. Up to April 2007, over 1,400 orphan products have been designated <strong>and</strong> a<br />

little more than 300 orphan products (of which 85% are drugs) have been approved<br />

since 1983. 37


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 29<br />

3.3.2 <strong>Orphan</strong> Drug Designation<br />

A sponsor can apply <strong>for</strong> the designation at the OOPD at anytime during the<br />

development process.<br />

The product has to satisfy one of the following criteria: 8<br />

1. prevalence criterion: less than 200,000 persons in the US suffer from the<br />

disease; or<br />

2. return on investment: even if there are more than 200,000 persons in the US<br />

suffer from the disease, there is no expectation that the costs of research <strong>and</strong><br />

developing of the drug <strong>for</strong> the indication can be recovered by sales of the<br />

drug in the US.<br />

The drug can also obtain the <strong>Orphan</strong> Designation if scientists <strong>and</strong> economists of the<br />

FDA determine that the drug will not be profitable <strong>for</strong> seven years after FDA approval,<br />

regardless of the number of patients affected.<br />

In May 2008 there were 325 designated orphan drugs with a marketing approval of<br />

which three were approved based on the ‘return on investment’-criterion. 40<br />

The <strong>Orphan</strong> Drug Designation will not be affected by a change in prevalence of the<br />

disease. 41<br />

In order to apply <strong>for</strong> an <strong>Orphan</strong> Drug Designation, the sponsor must submit following<br />

elements to the OOPD: 42<br />

• a statement requesting <strong>Orphan</strong> Drug Designation <strong>for</strong> a rare disease or<br />

condition;<br />

• a description of the rare disease or condition, the proposed indication or<br />

indications <strong>for</strong> use of the drug, <strong>and</strong> the reasons why such therapy is<br />

needed;<br />

• a description of the drug <strong>and</strong> a discussion of the scientific rationale <strong>for</strong> the<br />

use of the drug <strong>for</strong> the rare disease or condition;<br />

• if an alternative already exists, an explanation why the proposed variation<br />

may be clinically superior to the first drug m ;<br />

• if the drug is intended <strong>for</strong> a subset of persons with a particular disease or<br />

condition, a demonstration that the subset is medically plausible;<br />

• a summary of the regulatory status <strong>and</strong> marketing history of the drug in<br />

the USA <strong>and</strong> <strong>for</strong>eign countries;<br />

• documentation confirming one of the two abovementioned criteria;<br />

The OOPD must <strong>for</strong>mulate an answer within 60 days following the request.<br />

The designation will give the sponsor benefits to develop a drug <strong>for</strong> a rare disease or<br />

condition. There are five incentives:<br />

• tax credit of 50% <strong>for</strong> costs of clinical research undertaken in the USA;<br />

• access to the OODP’s clinical research grants program (even if the<br />

designation has not yet be obtained)<br />

(http://www.fda.gov/orphan/grants/index.htm);<br />

• marketing exclusivity <strong>for</strong> 7 years;<br />

• waiver of FDA user fees (always granted in the US. In the EU this in on<br />

request in EU <strong>and</strong> only <strong>for</strong> 50%);<br />

• Development <strong>and</strong> Regulatory assistance.<br />

m Clinically superior is defined as having greater effectiveness or greater safety in a substantial portion of<br />

the target population or demonstration that the drug makes a major contribution to patient care (FDA’s<br />

orphan drug regulations (21 C.F.R. Part 316))


30 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Other support measures are:<br />

• Compassionate use<br />

• Fast track approval <strong>for</strong> a drug if (1) the condition is serious or lifethreatening<br />

<strong>and</strong> (2) there is an unmet medical need <strong>for</strong> this condition.<br />

This measure does not exist as such in the EU: an expedited review or<br />

accelerated marketing approval is possible.<br />

• Paediatric developments are exempted from user fees if they meet the<br />

criteria: paediatric patients constitute a medically plausible subset of<br />

patient population.<br />

Unlike in the EU, there is no guidance <strong>for</strong> clinical trials in small populations.<br />

A new similar drug can not obtain the same <strong>Orphan</strong> Designation unless it proves to<br />

have a clinical superiority. Clinically superior means that a drug is shown to provide a<br />

significant therapeutic advantage over <strong>and</strong> above that provided by an approved orphan<br />

drug (that is otherwise the same drug) by showing a greater effectiveness, a greater<br />

safety or making a major contribution to patient care. 38<br />

The marketing exclusivity is granted <strong>for</strong> seven years (instead of ten years in the EU), but<br />

this can be shortened if one of the following criteria applies (Section 316.31 of 21<br />

CFR): 38<br />

• the <strong>Orphan</strong> Designation is withdrawn or revoked by the FDA;<br />

• the marketing approval is withdrawn;<br />

• the sponsor having the exclusive approval agrees with the withdrawal;<br />

• or the sponsor can not provide sufficient quantity of the drug.<br />

Figure 3.10 : <strong>Orphan</strong>-drug designations – by calendar year<br />

140<br />

120<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

53<br />

Number of approved <strong>Orphan</strong> Drug Designations 2000-2006<br />

87<br />

72<br />

110<br />

121 122 122<br />

2000 2001 2002 2003 2004 2005 2006<br />

[Source]: Based on Lewis DY. FDA Office of <strong>Orphan</strong> Products Development - Update 2007.<br />

NORD Corporate Council Meeting 21 May 2007. Available from<br />

[Last accessed: 11/12/2008].<br />

The sponsor will within 14 months after the designation date <strong>and</strong> annually thereafter<br />

until marketing approval submit a brief progress report to the OOPD (Section 316.30<br />

of 21 CFR) (as in the EU). 38<br />

EMEA <strong>and</strong> FDA have developed a common EU/US <strong>Orphan</strong> Drug Application, but the<br />

assessments of both agencies remain different.<br />

New is the paediatric drug development. A paediatric indication may be considered an<br />

orphan indication: the same criteria <strong>and</strong> incentives apply. But the incentives can only be<br />

used <strong>for</strong> the clinical paediatric drug development <strong>and</strong> the marketing approval can only<br />

be used <strong>for</strong> a paediatric indication.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 31<br />

3.3.3 Marketing approval<br />

A marketing approval is compulsory <strong>for</strong> a drug to be distributed or transported across<br />

the United States. As this may be necessary <strong>for</strong> clinical testing of a new drug or to<br />

provide treatment with a drug showing positive results during clinical testing, the<br />

sponsor can obtain an Investigational New Drug (IND). n Once the IND is approved by<br />

the FDA, clinical trials can start.<br />

The Marketing Approval is obtained by applying <strong>for</strong> a New Drug Application. The<br />

evaluation of the application is per<strong>for</strong>med by the Centre <strong>for</strong> Drug Evaluation <strong>and</strong><br />

Research (CDER) of the FDA <strong>and</strong> will last <strong>for</strong> ten months. Following elements are to be<br />

included in the submission file: 43<br />

• non-clinical studies;<br />

• clinical studies;<br />

• CMC in<strong>for</strong>mation: chemistry, manufacturing <strong>and</strong> controls;<br />

• proposed labelling;<br />

• additional in<strong>for</strong>mation.<br />

The drug will be approved if it demonstrates clinical benefit through clinical trials.<br />

There is an accelerated approval of new drugs <strong>for</strong> serious or life-threatening diseases:<br />

these drugs provide meaningful therapeutic benefit to patients over existing<br />

treatments. 44<br />

3.3.4 Compassionate use<br />

Compassionate use is a method of providing experimental therapeutics prior to final<br />

marketing approval <strong>for</strong> use in humans. This procedure is used with very sick individuals<br />

who have no other treatment options. A Treatment Investigational New Drug (t-IND)<br />

can be obtained regarding some conditions:<br />

• drug must be intended <strong>for</strong> the treatment of a serious or life-threatening<br />

disease;<br />

• no alternative drug of treatment must be available;<br />

• the product must be in the process of clinical trials <strong>and</strong> in an active phase<br />

of MA.<br />

3.4 EMEA-FDA COMPARISON<br />

The table below outlines the main differences in EMEA <strong>and</strong> FDA procedures governing<br />

<strong>Orphan</strong> Designation <strong>and</strong> Marketing Authorisation of orphan drugs.<br />

When considering applications <strong>for</strong> <strong>Orphan</strong> Designation, EMEA focuses on the health<br />

impact of the disease, its prevalence <strong>and</strong> treatment approaches. Although FDA also<br />

examines the prevalence of the disease, this is mainly considered from the perspective<br />

of estimating the return on investment on developing a drug <strong>for</strong> the indication.<br />

Furthermore, FDA applies a higher maximum prevalence <strong>for</strong> a disease to be designated<br />

as an orphan indication than EMEA. Unlike EMEA, the FDA does not allow<br />

reconsideration of an application <strong>for</strong> an <strong>Orphan</strong> Designation.<br />

Market exclusivity is an incentive to entice pharmaceutical companies to develop orphan<br />

drugs as the drug would otherwise not offer a return on investment due to the low<br />

prevalence of the indication. EMEA has in place a longer period of market exclusivity<br />

than FDA. However, the European Commission can shorten this period on the request<br />

of a member state .<br />

Both EMEA <strong>and</strong> FDA have incentives in place to apply <strong>for</strong> <strong>Orphan</strong> Drug Designation by<br />

offering the possibility to reduce or waive application fees; by offering assistance in<br />

preparing the application file; <strong>and</strong> by offering access to an accelerated review procedure.<br />

n http://www.fda.gov/cder/regulatory/applications/ind_page_1.htm


32 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

In the USA, a tax reduction on clinical studies of orphan drugs can be granted, whereas<br />

in Europe, various financial incentives are available on a national basis.<br />

EMEA has developed specific guidance <strong>for</strong> clinical trials in small populations, which is<br />

particularly relevant to the case of rare diseases <strong>and</strong> to paediatric orphan drugs. No<br />

such guidance has been issued by the FDA.<br />

In Europe, some countries have in place procedures governing compassionate use of<br />

orphan drugs. These procedures are outlined in the Chapter “Benchmarking of rare<br />

disease <strong>and</strong> drug markets in Europe”.<br />

45 46<br />

Figure 3.11: Comparison of EMEA <strong>and</strong> FDA in the field of <strong>Orphan</strong> <strong>Drugs</strong><br />

EMEA FDA<br />

<strong>Orphan</strong> Designation - Life-threatening or chronically - Less than 200,000 persons in<br />

Criteria<br />

debilitating diseases, with the USA; or there is no<br />

prevalence < five per 10,000 expectation that drug R&D costs<br />

population; or life-threatening, <strong>for</strong> the indication can be<br />

seriously debilitating or serious recovered by sales in USA<br />

<strong>and</strong> chronic condition where, - if FDA determines that drug will<br />

without incentives, there not be profitable <strong>for</strong> seven years<br />

would be no justification <strong>for</strong> after FDA approval, regardless of<br />

investing in development of<br />

treatment<br />

- No satisfactory treatment<br />

should exist or product must<br />

be of significant benefit to<br />

those with condition<br />

number of patients affected<br />

Reconsideration of<br />

<strong>Orphan</strong> Designation<br />

application<br />

Yes, every 6 months No<br />

Prevalence Fewer than 5 per 10,000 Fewer than 6.6 per 10,000<br />

Institution in charge Committee of <strong>Orphan</strong> Medicinal<br />

Products<br />

Marketing<br />

Authorisation<br />

- Application <strong>for</strong> orphan<br />

medicinal product designation<br />

- Marketing Authorisation<br />

application <strong>for</strong> orphan drug<br />

Market exclusivity 10 years (12 years <strong>for</strong> paediatric<br />

drugs)<br />

Research funding Money from national authorities &<br />

Community grants<br />

Private sources<br />

Financial incentives Financial incentives on a national<br />

basis<br />

Maximum more or less 250,000<br />

patients affected or financially nonviable<br />

Fee waiver via request: given by<br />

some Member States <strong>and</strong> by EMEA<br />

<strong>for</strong> centralised applications<br />

Office of <strong>Orphan</strong> Products<br />

Development<br />

- Ask orphan drug status -<br />

OOPD<br />

- Ask Marketing Authorisation<br />

– one of the Centres of FDA<br />

7 years<br />

Money by National Institutes of<br />

Health programmes <strong>and</strong> others<br />

Private sources<br />

Tax reduction: 50% <strong>for</strong> clinical<br />

studies<br />

Maximum 200,000 patients<br />

affected or financially non-viable<br />

Always fee reduction<br />

Assistance with Development <strong>and</strong> Regulatory Development <strong>and</strong> Regulatory


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 33<br />

application file<br />

Accelerated marketing<br />

procedure<br />

EMEA FDA<br />

assistance assistance<br />

Possible access to accelerated<br />

review<br />

Small populations Guidance <strong>for</strong> clinical trials in small<br />

populations<br />

Compassionate use - Procedure at EMEA level <strong>for</strong><br />

medicinal products not yet<br />

having received a Marketing<br />

Authorisation<br />

- Procedure on a national level<br />

different per member state<br />

Access to fast-track<br />

No guidance <strong>for</strong> clinical trials in<br />

small populations<br />

A Treatment Investigational New<br />

Drug (t-IND) can be obtained


34 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

4 INTERNATIONAL COMPARISON OF RARE<br />

DISEASE AND DRUG MARKETS IN EUROPE<br />

4.1 INTRODUCTION<br />

The previous chapter has described the EMEA procedure from <strong>Orphan</strong> Designation to<br />

Marketing Authorisation <strong>and</strong> compared this with the FDA procedure in the United<br />

States. The present chapter describes the regulatory aspects of the rare disease <strong>and</strong><br />

drug market in Belgium <strong>and</strong> in a number of other countries (i.e. France, Italy, the<br />

Netherl<strong>and</strong>s, Sweden <strong>and</strong> the United Kingdom). A description of the regulation in each<br />

country is followed by a comparative analysis between these countries.<br />

Key points<br />

• This chapter examines regulatory aspects of rare disease <strong>and</strong> drug markets<br />

in a number of countries;<br />

• Regulation of the Belgian market is compared with regulation governing the<br />

Dutch, French, Italian, Swedish <strong>and</strong> British markets.<br />

4.2 METHODOLOGY<br />

First, the rare disease <strong>and</strong> drug market has been described <strong>for</strong> Belgium, France, Italy, the<br />

Netherl<strong>and</strong>s, Sweden, <strong>and</strong> the United Kingdom. Second, a comparative analysis was<br />

made focussing on different relevant aspects such as the institutional context, the<br />

national Marketing Authorisation procedures, pricing, reimbursement procedures,<br />

distribution channels <strong>and</strong> prescribing processes. Various elements of the institutional<br />

context were explored, including centres <strong>for</strong> rare diseases <strong>and</strong>/or orphan drugs, policy<br />

measures supporting the development of orphan drugs, <strong>and</strong> incentives <strong>for</strong> research on<br />

rare diseases <strong>and</strong>/or orphan drugs. Additionally, the analysis enquired about the criteria<br />

<strong>for</strong> compassionate use <strong>and</strong> off-label use <strong>for</strong> orphan drugs. Pricing issues related to<br />

whether a country has a system of free o or fixed p pricing of orphan drugs. If fixed pricing<br />

exists, the criteria <strong>for</strong> price setting were examined. The mechanism <strong>for</strong> reimbursing<br />

orphan drugs <strong>and</strong> whether orphan drugs are fully or partially reimbursed is also<br />

covered. In<strong>for</strong>mation was gathered about the distribution channels <strong>and</strong> site of delivery<br />

of orphan drugs. A final issue concerned the conditions <strong>for</strong> prescribing orphan drugs.<br />

The countries were selected <strong>for</strong> their comparable living st<strong>and</strong>ards <strong>and</strong> their geographic<br />

proximity to Belgium. Furthermore, the chosen country panel provides insight into the<br />

variety of regulatory mechanisms that govern rare disease <strong>and</strong> drug markets. Finally,<br />

health expenditure is primarily financed by the public payer (the third-party payer or<br />

National Health Service) in each of these countries. 47<br />

A review of the international peer-reviewed literature confirmed the absence of<br />

scientific articles on the regulation of rare disease <strong>and</strong> drug markets. There<strong>for</strong>e,<br />

in<strong>for</strong>mation was gained by accessing documents setting out national legislation <strong>and</strong> local<br />

publications. In addition, a qualitative questionnaire (see annex 1.3) was completed by<br />

correspondents from governmental <strong>and</strong> regulatory agencies, rare disease <strong>and</strong> orphan<br />

drug national task <strong>for</strong>ces, patient organisations, health insurance funds <strong>and</strong> members of<br />

the INAHTA (International Network of Agencies <strong>for</strong> Health Technology Assessment) q .<br />

Members of the COMP at EMEA <strong>and</strong> the European Task Force on <strong>Rare</strong> <strong>Diseases</strong> also<br />

provided in<strong>for</strong>mation.<br />

o In a free price system, the price is set following negotiation between the government <strong>and</strong> the sponsor.<br />

p In a fixed price system, the price level is fixed by the government; no deviation by the sponsor is possible.<br />

q http://www.inahta.org/inahta_web/index.asp


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 35<br />

The data collection relied on a literature search, a survey based on a qualitative<br />

questionnaire <strong>and</strong> telephone interviews with national experts. Regarding Belgium, a<br />

meeting took place with Pharma.be, the representative organ of the Belgian<br />

pharmaceutical industry in order to comprehend their point of view of the Belgian<br />

situation.<br />

Each country-specific section about the regulation governing rare diseases <strong>and</strong> drugs<br />

was validated by a national expert.<br />

Key points<br />

• The benchmarking exercise focused on issues related to the institutional<br />

context, Marketing Authorisation, reimbursement, pricing, distribution <strong>and</strong><br />

prescription of orphan drugs.<br />

• Regulatory aspects of rare disease <strong>and</strong> drug markets were examined<br />

through the perusal of legal texts, the analysis of survey results, <strong>and</strong> contacts<br />

with national experts.<br />

4.3 BELGIUM<br />

4.3.1 Institutional context<br />

Belgium applies the EU definition <strong>for</strong> rare diseases. The main institutional actor in the<br />

Belgian orphan drug policy is the National Institute <strong>for</strong> Health <strong>and</strong> Disability Insurance<br />

(NIHDI). There are no official centres of reference <strong>for</strong> rare diseases, but there are<br />

several centres that are specialised in one or more rare diseases. Some of these centres<br />

are recognised by the NIHDI <strong>and</strong> work under a convention. These centres include the<br />

eight centres <strong>for</strong> human genetics 48 , the seven Mucoviscidose (CF) centres, eleven<br />

centres <strong>for</strong> metabolic diseases <strong>and</strong> six <strong>for</strong> neuro-muscular diseases. The NIHDI has<br />

restricted the reimbursement of some orphan drugs to prescribers belonging to one of<br />

the recognised centres that provide treatment.<br />

No specific programmes to fund research networks exist. There are not yet national<br />

policy measures to promote the development of orphan drugs, although there is a<br />

growing dem<strong>and</strong> on the part of patients, the medical community <strong>and</strong> even politicians.<br />

Still, revenues of orphan drugs are not subject to taxation. A Pilot Group <strong>for</strong> <strong>Orphan</strong><br />

<strong>Drugs</strong> (Stuurgroep Weesgeneesmiddelen) 49 was established in order to promote a<br />

coherent policy <strong>for</strong> orphan drugs <strong>and</strong> rare diseases. The Pilot Group comprises<br />

different thematic working groups composed of experts of different horizons. Discussed<br />

themes are survey & registries, rare diseases & costs, in<strong>for</strong>mation & education, <strong>and</strong><br />

reference centres. The Pilot Group has been integrated in the Fund <strong>Rare</strong> <strong>Diseases</strong> <strong>and</strong><br />

<strong>Orphan</strong> <strong>Drugs</strong> of the King Baudouin Foundation.<br />

4.3.2 Marketing Authorisation<br />

<strong>Orphan</strong> drugs obtain Marketing Authorisation through the centralised procedure at<br />

EMEA since 2005 (see previous chapter). Be<strong>for</strong>e 2005, Marketing Authorisation through<br />

the mutual recognition procedure was till possible. In Belgium this has been the case <strong>for</strong><br />

e.g. Duodopa.


36 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

4.3.3 Reimbursement<br />

In order <strong>for</strong> a drug to be put on the Belgian reimbursable pharmaceutical products lists,<br />

the Marketing Authorisation Holder (MAH) has to submit a drug reimbursement<br />

request to the Drug Reimbursement Committee (DRC) of the NIHDI. Mid-February<br />

2009, there 39 application files had been submitted of which 36 had been examined. 32<br />

applications received a positive advice <strong>and</strong> four a negative r . 50 Three files were ongoing at<br />

that time. An application <strong>for</strong> reimbursement in Belgium can be submitted once the<br />

CHMP has given a positive advice, 51 but companies will do this most often only after the<br />

marketing authorisation was obtained.<br />

Figure 4.1 : Composition of the Drug Reimbursement Committee<br />

The Drug Reimbursement Committee is composed of 28 members: 1<br />

• 22 voting members:<br />

o 7 academics<br />

o 8 representatives of the sickness funds<br />

o 4 representatives of the physicians’ association<br />

o 3 representatives of the pharmacists association<br />

• 6 non voting members<br />

o 3 ministry representatives (Ministry of Health, of Social Affairs <strong>and</strong><br />

of Economical Affairs)<br />

o 1 representative of the NIHDI<br />

o 2 members of Pharma.be (which is the representative organisation<br />

of the pharmaceutical industry in Belgium)<br />

If a medicinal product is not yet on the Belgian reimbursable pharmaceutical products<br />

lists, the patient can apply <strong>for</strong> compassionate use (see point 4.3.3.2) or <strong>for</strong><br />

reimbursement through the Special Solidarity Fund (see point 4.3.3.3).<br />

4.3.3.1 Application procedure <strong>for</strong> reimbursement<br />

To be added on the Belgian reimbursable pharmaceutical products lists, orphan drugs<br />

follow the same procedure as the drugs of class 1 <strong>and</strong> others s (being specialties <strong>for</strong><br />

which the company claims added therapeutic value in comparison to therapeutic<br />

alternatives) (see article 5 Royal Decree 21/12/2001) 52 , but are considered to be a<br />

specific category of drugs within class I. The procedure is the same, but the requested<br />

in<strong>for</strong>mation is different. For example, in contrast to class I pharmaceutical products,<br />

drug reimbursement request files <strong>for</strong> orphan drugs do not have to include a costeffectiveness<br />

analysis.<br />

The pharmaceutical company introduces two dossiers: an application <strong>for</strong>m sent to the<br />

secretariat of the DRC <strong>and</strong> a price dem<strong>and</strong> to the Federal Public Service (FPS)<br />

Economy t (see point 1.3.4 of this chapter).<br />

r This is the situation at that specific date <strong>and</strong> includes potentially multiple applications <strong>for</strong> a same drug (e.g.<br />

refused, then resubmitted <strong>and</strong> either approved or not).<br />

s There are three added value classes in Belgium. The therapeutic value of a medicinal product is decided<br />

by the DRC <strong>and</strong> expressed in an added value class.<br />

Class I: medicinal products of which the therapeutic added value has been proved compared to existing<br />

therapeutic alternatives;<br />

Class 2: medicinal products with no proven therapeutic added value compared to existing therapeutic<br />

alternatives;<br />

Class 3: other medicinal products – categorised according to legislation.<br />

t FPS st<strong>and</strong>s <strong>for</strong> Federal Public Service known <strong>for</strong>merly as Ministry.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 37<br />

Once the dossier has been received by the DRC, a period of 180 days starts within<br />

which a reimbursement decision has to be taken. The dossier must contain three types<br />

of data (art. 37, RD 21/12/2001):<br />

• the indication of the orphan drug as set by the Community register of<br />

orphan medicinal products <strong>and</strong> the important motivations on which the<br />

approval was based; 53<br />

• a copy of the dem<strong>and</strong> sent to the FPS Economy;<br />

• a proposal regarding the reimbursement level <strong>and</strong> a justification thereof<br />

(including therapeutic value, budgetary impact <strong>and</strong> therapeutic <strong>and</strong> social<br />

needs) 52 .<br />

The DRC may decide to compose a group of experts to evaluate the justification of the<br />

reimbursement proposal. Even if not, a first temporary evaluation report will be<br />

elaborated by the DRC (together with the experts) <strong>and</strong> sent to the company within 30<br />

days. The final evaluation report is sent within 60 days of the dossier introduction. The<br />

company has 20 days to <strong>for</strong>ward objections <strong>and</strong> remarks to the DRC, or to ask <strong>for</strong><br />

more time to respond. (art. 15§1, RD 21/12/2001)<br />

After having received the company’s answers, the DRC prepares a temporary proposal<br />

(containing the added value class (i.e. class 1), the reimbursement conditions, the<br />

reimbursement base, the reimbursement category <strong>and</strong> the revision criteria) <strong>for</strong> drug<br />

reimbursement u if the proposal differs from the company’s proposal. Otherwise the<br />

DRC will prepare a final proposal <strong>and</strong> this within a period of 150 days following<br />

reception of the application.<br />

u This proposal will be included in Chapter IV of the Royal Decree of 21/12/2001.


Day 0<br />

Day 8<br />

Day 30<br />

Day 60<br />

38 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Day 120<br />

Day 150<br />

Day 180<br />

Figure 4.2 : Procedure <strong>for</strong> the inclusion of an orphan drug on the drug<br />

reimbursement list<br />

Drug Reimbursement Committee FPS Economy<br />

Pharmaceutical company submits appli-cation<br />

<strong>for</strong>m to the secretariat of DRC<br />

Admissibility check by secretariat: <strong>for</strong>m is<br />

sent over to Bureau of DRC<br />

First scientific report written by experts <strong>and</strong><br />

the Bureau of DRC<br />

Evaluation report written by experts in<br />

dialogue with the DRC<br />

Company receives report: can <strong>for</strong>mulate<br />

remarks <strong>and</strong> answer DRC’s questions<br />

DRC prepares temporary proposal <strong>for</strong> drug<br />

reimbursement <strong>and</strong> in<strong>for</strong>ms company<br />

Company must react within 10 days<br />

DRC prepares final proposal with reimbursement<br />

conditions <strong>and</strong> revision criteria<br />

Pharmaceutical company introduces price<br />

dem<strong>and</strong> at FPS Economy<br />

FPS Economy decides on the maximum price<br />

<strong>for</strong> the company to h<strong>and</strong>le<br />

Notification of price to company<br />

Company has to in<strong>for</strong>m CTG of maximum<br />

price within 90 days following the<br />

decision on price dem<strong>and</strong><br />

Company Minister of Social Affairs Advice of Finance inspector <strong>and</strong><br />

approval Minister of Budget<br />

Minister takes final decision <strong>and</strong> in<strong>for</strong>ms company<br />

FPS Budget<br />

The inclusion on the reimbursement list is approved when the drug has a certain<br />

therapeutic value, being the sum of the evaluation of all the speciality’s properties<br />

relevant <strong>for</strong> the treatment: this is the efficacy, the usefulness, the tolerance, the<br />

applicability <strong>and</strong> the user friendliness. Together, these elements determine the place of<br />

the speciality within the therapy compared to other available treatments. The<br />

therapeutic value is situated at the level of morbidity, mortality <strong>and</strong> quality of life. A<br />

speciality has a therapeutic added value if the treatment with the concerned speciality<br />

has a higher therapeutic value than the recognized st<strong>and</strong>ard treatment.” 52<br />

Day 0<br />

Day 90


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 39<br />

The Minister of Social Affairs will take the final decision within 180 days: he or she is<br />

not bound by the DRC’s advice <strong>and</strong> can take a different decision. The Minister will ask<br />

the advice of the Inspector of Finance <strong>and</strong> receive the approval of the Minister of<br />

Budget. The Minister of Social Affairs will always follow the advice of the Minister of<br />

Budget. 50 This process is taking place during the last 30 days of the process leaving little<br />

time <strong>for</strong> negotiation. The approval of the Minister of Budget is needed because of<br />

budgetary implications giving a de facto veto right on budgetary grounds.<br />

Current situation<br />

On the 31 st of December 2008, 31 orphan drugs were reimbursed in Belgium <strong>for</strong> a total<br />

of 35 orphan indications v . This includes Glivec® that is approved as a Class II drug (not<br />

as a Class I as all other orphan drugs). There are several non-orphan drugs that are<br />

reimbursed <strong>for</strong> orphan indications although they do not have the Emea MA.<br />

Figure 4.3 : Total number of reimbursed orphan drugs in Belgium 1999-2008<br />

Number of new <strong>Orphan</strong> <strong>Drugs</strong> per year<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

1999 2003 2004 2005 2006 2007 2008<br />

Number of new <strong>Orphan</strong> <strong>Drugs</strong> per year Total number of <strong>Orphan</strong> <strong>Drugs</strong><br />

Source: NIHDI. Farmaceutische specialiteiten.<br />

. Accessed 2008-2009,<br />

1/3/2009.<br />

Of these 31 drugs, 30 have obtained a marketing authorisation at EU level. This means<br />

that as of 31 December 2008, there were 17 orphan drugs with a MA that were not<br />

reimbursed in Belgium. Of these 17 drugs:<br />

• two have been approved since then (in 2009)<br />

• two have been refused (Pedea <strong>and</strong> Wilzin)<br />

• one drug has probably not been submitted as there is an alternative on<br />

the Belgian market (Siklos, as Hydrea is on the market)<br />

Of the twelve drugs left, 3 received their MA in 2008, <strong>and</strong> one can expect the<br />

manufacturers will request reimbursement in Belgium during 2009.<br />

<strong>Orphan</strong> drugs within the therapeutic area endocrinology/metabolism account <strong>for</strong> the<br />

largest share in the total number of orphan drugs (35% of all orphan drugs are <strong>for</strong><br />

endocrinology/metabolic conditions), followed closely by the oncology drugs (31%).<br />

Savene®, <strong>for</strong> the treatment of anthracycline extravasations, is included in the “other”<br />

category.<br />

v Please refer to table 9.1 in annex <strong>for</strong> the full list of orphan drugs.<br />

35<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

Total number of <strong>Orphan</strong> <strong>Drugs</strong>


40 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Figure 4.4 : Reimbursed Belgian <strong>Orphan</strong> drugs by therapeutic area<br />

Cardiovascular<br />

& respiratory<br />

9%<br />

Haematology<br />

19%<br />

Nervous<br />

system<br />

3%<br />

Other<br />

3% Oncology<br />

31%<br />

Endocrinology /<br />

metabolism<br />

35%<br />

Source: EMEA. List of orphan-designated authorised medicines 6/11/2008. Available from<br />

[Last accessed: 7/5/2009].<br />

4.3.3.2 Compassionate use <strong>and</strong> Belgian Medical Need programme<br />

The Law of 1/5/2006 54 provides <strong>for</strong> compassionate use, this is the treatment with drugs<br />

which are not yet reimbursed or available in Belgium. There are two programmes:<br />

• Programmes of compassionate use: making available, <strong>for</strong> compassionate<br />

reasons, of a medicinal product that can qualify <strong>for</strong> the centralised<br />

procedure to a group of patients with a chronically or seriously<br />

debilitating disease or whose disease is considered to be life-threatening,<br />

<strong>and</strong> who cannot be treated satisfactorily by an authorised medicinal<br />

product. The medicinal product concerned must either be the subject of<br />

an application <strong>for</strong> a Marketing Authorisation in accordance with Article 6<br />

of the European Regulation or must be undergoing clinical trials. 55<br />

• The Medical Need Programmes: making available a medicinal product to a<br />

group of patients with a chronically or seriously debilitating disease or<br />

whose disease is considered to be life-threatening, <strong>and</strong> who can not be<br />

treated satisfactorily by an authorised medicinal product. The medicinal<br />

product concerned must have a Marketing Authorisation but<br />

o either the given indication has not been authorised yet, or<br />

o although authorised, the medicinal product is not yet available<br />

on the market in this indication. 55<br />

The essential difference between the two programmes is that Compassionate Use<br />

concerns medicinal products which do not yet have obtained a Marketing Authorisation<br />

in Belgium, unlike the Medical Need Programme, which concerns medicinal products<br />

which have a Marketing Authorisation in Belgium <strong>for</strong> a given indication.<br />

In order <strong>for</strong> a medicinal product to be considered <strong>for</strong> compassionate use, the MAH will<br />

have to introduce a dem<strong>and</strong> that will be reviewed <strong>and</strong> approved by one of the Belgian<br />

ethics committees. The compassionate use treatment will be prescribed by a physician:<br />

the hospital can require approval <strong>for</strong> the individual patient by the local ethics<br />

committee.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 41<br />

4.3.3.3 Special Solidarity Fund<br />

A patient can request reimbursement of an orphan drug or treatment unavailable w in<br />

Belgium through the Special Solidarity Fund (SSF), part of the NIHDI. The objective of<br />

the SSF is the reimbursement of medical expenses <strong>for</strong> rare diseases, rare indications <strong>and</strong><br />

innovative techniques which are not (yet) refunded by the compulsory health insurance.<br />

The legal base is the law of 27 April 2005. 56<br />

Treatment costs <strong>for</strong> rare indications <strong>and</strong> diseases can be reimbursed if a number of<br />

criteria are fulfilled (see table below). Reimbursement will only be granted if the patient<br />

has been through all other reimbursement options, including all applicable legislation at<br />

national, European or international level. This means that reimbursement through the<br />

SSF cannot be obtained if the reimbursement of the orphan drug has been refused by<br />

the CMDOD.<br />

Figure 4.5 : SSF’s Reimbursement Criteria<br />

Type of reimbursement Reimbursement criteria<br />

Reimbursement of treatment The treatment is expensive.<br />

costs <strong>for</strong> rare indications Medical treatment is prescribed by a medical doctor specialised in<br />

(art. 25 bis)<br />

the treatment of the related disorder <strong>and</strong> authorised to practice<br />

medicine in Belgium.<br />

Medical treatment has a scientific value <strong>and</strong> effectiveness which is<br />

largely recognized by the medical profession. The medical<br />

treatment has to have outgrown the experimental phase.<br />

The compulsory health insurance system cannot provide an<br />

alternative.<br />

Medical treatment is used <strong>for</strong> an indication threatening vital<br />

functions of the patient.<br />

Reimbursement of<br />

The medical treatment is considered as being expensive.<br />

treatments costs <strong>for</strong> rare The compulsory health insurance system does not provide a<br />

diseases (art. 25 ter § 1) therapeutic alternative treatment.<br />

Medical treatment is used <strong>for</strong> a rare disease that threatens the<br />

vital functions of the patient.<br />

The medical treatment is prescribed by a medical doctor<br />

specialized in the treatment of the specific disease <strong>and</strong> authorised<br />

to practice medicine in Belgium.<br />

The medical profession recognizes the treatment as the specific<br />

approach <strong>for</strong> the rare disease.<br />

Reimbursement of cost <strong>for</strong> Treatment as a whole is expensive.<br />

rare diseases requiring a Treatment is related to a threat of the vital functions of a patient.<br />

continuous <strong>and</strong> complex The threat of the vital functions is directly <strong>and</strong> specifically a<br />

treatment (art. 25 ter § 2) consequence of the rare disease.<br />

The compulsory health insurance system does not provide<br />

therapeutic alternative.<br />

The complex treatment is prescribed by a medical doctor,<br />

specialized in the treatment of the specific disease <strong>and</strong> authorised<br />

to practice medicine in Belgium.<br />

Source: NIHDI. Het Bijzonder Solidariteitsfonds. Wat is de functie ervan? Wanneer en hoe kunt u<br />

er een beroep op doen? 2006. Available from<br />

[Last accessed:<br />

14/4/2008].<br />

w Whether or not this orphan drug has a MA or is reimbursed abroad.


42 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

The NIHDI body responsible <strong>for</strong> managing the SSF <strong>and</strong> taking decisions about<br />

reimbursements is the College of Medical Doctors Directors composed of Medical<br />

Doctor Directors of each national sickness fund <strong>and</strong> of NIHDI MDs.<br />

The patient’s Medical Doctor (MD) (a specialist) will fill out a <strong>for</strong>m <strong>and</strong> h<strong>and</strong> it over to<br />

the health insurance institute through the local <strong>and</strong> national sickness fund. Within one<br />

month, the College of MD directors will examine the application <strong>and</strong> take a decision.<br />

The decision is transmitted to the patient <strong>and</strong> to the local sickness fund. In case of a<br />

positive advice, this latter will proceed to the reimbursement within fifteen days. The<br />

request <strong>for</strong> reimbursement must be done within three years following the end of the<br />

treatment.<br />

Figure 4.6 : Special Solidarity Fund Procedure<br />

Introduction<br />

reimbursement<br />

dem<strong>and</strong><br />

Patient<br />

Local Sickness Fund<br />

National Sickness Fund<br />

NIHDI/SSF – College of MD<br />

Directors<br />

Reimbursement<br />

decision<br />

NIHDI = National Institute <strong>for</strong> Health <strong>and</strong> Disability Insurance; SSF = Special Solidarity Fund<br />

The SSF has in 2007 reimbursed five drugs with <strong>Orphan</strong> Designation (not yet<br />

reimbursed as orphan drugs) <strong>for</strong> 141 patients <strong>for</strong> a total amount of € 4 084 225 (€<br />

28 966 per patient). This accounts <strong>for</strong> 35% of the SSF’s budget. The table below gives an<br />

overview of these five drugs. From the five products, five have been approved <strong>for</strong><br />

reimbursement as orphan drugs since.<br />

Figure 4.7 : SSF reimbursement <strong>for</strong> <strong>Orphan</strong> <strong>Drugs</strong> with MA in 2007<br />

<strong>Orphan</strong> drug Total NIHDI Number of patients NIHDI expenditures<br />

Expenditures<br />

per patient<br />

Myozyme® € 3 540 723 7 € 505 818<br />

Revatio® € 299 358 67 € 4 468<br />

Revlimid® € 141 050 57 € 2 475<br />

Ventavis/Iloprost® € 100 927 9 € 11 214<br />

Tracleer® € 2 167 1 € 2 167<br />

Source: NIHDI. Jaarverslag 2007 betreffende het Bijzonder Solidariteitsfonds, 2008


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 43<br />

4.3.4 Pricing<br />

The pharmaceutical company introduces a price dem<strong>and</strong> at the Federal Public Service<br />

(FPS) Economy x at the same time a reimbursement dem<strong>and</strong> is introduced. This<br />

application consists of 51<br />

• name <strong>and</strong> address of the MAH;<br />

• name, pharmaceutical <strong>for</strong>m, accurate indication <strong>and</strong> (if applicable)<br />

therapeutic added value of the drug;<br />

• a statement of registration proof, the scientific instruction leaflet <strong>and</strong> the<br />

public instruction leaflet;<br />

• a justification <strong>for</strong> the proposed price in terms of cost drivers;<br />

• the annual accounts of the applicant <strong>for</strong> the last three years;<br />

• the market <strong>and</strong> competition conditions <strong>and</strong> a price comparison with other<br />

EU Member States.<br />

The in<strong>for</strong>mation to be provided <strong>for</strong> orphan drugs is the same as <strong>for</strong> non orphan drugs.<br />

Still, the evaluation might differ according to the actual in<strong>for</strong>mation provided as there<br />

are no st<strong>and</strong>ard reporting requirements <strong>for</strong> costs imposed.<br />

The FPS Economy compares the given in<strong>for</strong>mation with available data (such as other<br />

drug dossiers, other countries <strong>and</strong> other similar therapeutic drugs). 50<br />

Within a period of 90 days the FPS Economy will decide on a price <strong>and</strong> in<strong>for</strong>m the<br />

company. The company must notify the price to the Drug Reimbursement Committee.<br />

The Committee takes that into account when making a reimbursement decision.<br />

No exchange of in<strong>for</strong>mation takes place between the FPS Economy <strong>and</strong> NIHDI during<br />

this period. Following the decision, the FPS Economy will play no other role but to<br />

collect the manufacturers’ turnover figures.<br />

4.3.5 Distribution<br />

Most orphan drugs, except <strong>for</strong> two (Glivec <strong>and</strong> Thalidomide) are distributed through<br />

the hospital pharmacies.<br />

4.3.6 Prescribing<br />

The prescription of orphan drugs is subject to conditions to be found in the<br />

reimbursement <strong>for</strong>m. <strong>Orphan</strong> drugs are part of the reimbursement category A y : these<br />

are drugs <strong>for</strong> severe conditions or diseases <strong>and</strong> are reimbursed at 100%. The conditions<br />

<strong>for</strong> reimbursement are described in the applications <strong>for</strong>ms <strong>for</strong> reimbursement to be<br />

found in Chapter IV of the Royal Decree of 21/12/2001. 52 This chapter contains all<br />

drugs that receive special reimbursement conditions due to medical <strong>and</strong>/or budgetary<br />

reasons.<br />

A physician wishing to prescribe an orphan drug to a patient has to follow the<br />

procedure set out in Figure 4.8 in order to obtain reimbursement <strong>for</strong> this patient. The<br />

physician must receive the approval of a Medical Advisor of the sickness fund. This<br />

procedure also applies to a number of non-orphan drugs <strong>and</strong> is hence not specific <strong>for</strong><br />

orphan drugs. However, in case of orphan drugs the medical advisor has the additional<br />

possibility to ask advice from the “College of Medical Doctors <strong>for</strong> <strong>Orphan</strong> <strong>Drugs</strong>”<br />

(CMDOD) if one exists <strong>for</strong> the drug. The Medical Advisor of the sickness fund can, but<br />

is not obliged to, request the advice of the CMDOD.<br />

x FPS st<strong>and</strong>s <strong>for</strong> Federal Public Service known <strong>for</strong>merly as Ministry.<br />

y This categories define the reimbursement level <strong>and</strong> are different to the Classes mentioned above, which<br />

are based on the therapeutic value. <strong>Orphan</strong> drugs are automatically classified into the highest<br />

reimbursement category (100%) <strong>and</strong> in Class I.


44 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Figure 4.8: Procedure to request reimbursement of orphan drug <strong>for</strong> an<br />

individual patient<br />

Compulsory:<br />

Application <strong>for</strong> an orphan<br />

drug treatment<br />

Optional:<br />

Ask <strong>for</strong> advice<br />

Specialist MD of the patient<br />

Medical advisor of the Sickness<br />

Fund<br />

“College of Medical Doctors <strong>for</strong><br />

<strong>Orphan</strong> <strong>Drugs</strong>”<br />

Source: FOD Sociale Zekerheid. Koninklijk besluit van 8 juli 2004 betreffende de vergoeding van<br />

weesgeneesmiddelen. Belgisch Staatsblad, 20/07/2004.<br />

Individual reimbursement advice is <strong>for</strong>mulated on a case by case basis by the CMDOD if<br />

their involvement is required by the reimbursement modalities of the orphan drug. It is<br />

the DRC that decides whether or not a College is established. At the end of April 2009,<br />

there were eighteen colleges <strong>for</strong> 31 orphan drugs.<br />

A College is composed of a president, four specialist MDs in the indication/disease <strong>and</strong><br />

four MDs member of the DRC <strong>and</strong> m<strong>and</strong>ated by a sickness fund. 20<br />

It is the specialist MD of the patient who completes an application <strong>for</strong>m <strong>for</strong> the orphan<br />

drug to be found in Chapter VI of the Royal Decree of 21/12/2001 52 , <strong>and</strong> who submits<br />

the application to the sickness fund. It is imperative that the MD is affiliated to a<br />

recognised centre or a hospital <strong>for</strong> a certain disease, e.g.:<br />

• <strong>for</strong> metabolic diseases : a Revalidation Centre <strong>for</strong> monogenetic hereditary<br />

metabolic diseases; z<br />

• <strong>for</strong> haematology: a Centre <strong>for</strong> Haematology linked to a hospital;<br />

• <strong>for</strong> cardiology-pulmonology: a hospital.<br />

The Medical Advisor of the sickness fund will examine the dem<strong>and</strong> <strong>and</strong> can decide to<br />

request the advice of the CMDOD. Even if the Medical advisor is not obliged to request<br />

the advice of the CMDOD of the concerned orphan drug, in practice he or she <strong>for</strong> each<br />

decision aa always asks <strong>for</strong> advice. 57 The CMDOD <strong>for</strong>mulates its advice based on the<br />

reimbursement criteria defined in Chapter VI of the Royal Decree of 21/12/2001.<br />

The dem<strong>and</strong> <strong>for</strong> individual reimbursement has to be introduced every year as<br />

reimbursement approval is only valid <strong>for</strong> a period of twelve months.<br />

z http://www.NIHDI.fgov.be/care/all/revalidatie/general-in<strong>for</strong>mation/contacts/pdf/7890.pdf<br />

aa The pharmaceutical industry claims that not all reimbursement dem<strong>and</strong>s are submitted to the CMDOD.<br />

There is no evidence to confirm this statement.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 45<br />

Key points<br />

• In Belgium, there are no specific centres of reference, policy measures,<br />

research incentives on rare diseases/orphan drugs.<br />

• <strong>Orphan</strong> drugs are registered through the EMEA centralised procedure only.<br />

Specific legislation governs compassionate use of orphan drugs <strong>and</strong> there<br />

exists a programme <strong>for</strong> medical needs.<br />

• <strong>Orphan</strong> drug maximum prices are fixed by the Federal Public Service<br />

Economy as is the case <strong>for</strong> all drugs, independently of the reimbursement<br />

decision.<br />

• The reimbursement procedure considers budget impact, but not costeffectiveness.<br />

Pharmaceutical companies do not have to submit a <strong>for</strong>mal<br />

cost-effectiveness analysis as part of a drug reimbursement request file <strong>for</strong><br />

an orphan drug. <strong>Orphan</strong> drugs are fully reimbursed by the NIHDI.<br />

• <strong>Orphan</strong> drugs are distributed through hospital pharmacies only.<br />

• The prescription of orphan drugs by specialist physicians is subject to<br />

approval of a Medical Advisor of the sickness fund <strong>and</strong> in practice based on<br />

the advice of a College of Medical Doctors <strong>for</strong> <strong>Orphan</strong> <strong>Drugs</strong>.<br />

4.4 FRANCE bb<br />

4.4.1 Institutional context<br />

Three institutions play a role with regard to orphan drugs on the French market: the<br />

French Agency <strong>for</strong> the Sanitary Security of Health Products (Afssaps - Agence Française<br />

de Sécurité Sanitaire des Produits de Santé), the High Health Authority (HAS – Haute<br />

Autorité de Santé), <strong>and</strong> the Ministry of Health. The HAS is a public independent<br />

authority having as objectives to improve the quality <strong>and</strong> security of the health services,<br />

to maintain a high-per<strong>for</strong>mance health system <strong>and</strong> to in<strong>for</strong>m patients on their diseases<br />

<strong>and</strong> treatment.<br />

The institution in charge of research on rare diseases is the GIS cc – Institut des Maladies<br />

rares whose objectives are to define <strong>and</strong> establish a national policy <strong>for</strong> research on rare<br />

diseases; to mobilise the competences <strong>and</strong> to enhance multidisciplinary approaches; <strong>and</strong><br />

to coordinate the research <strong>and</strong> to associate existing means. 58<br />

Measures taken to promote the orphan drug policy of the EU:<br />

• 2001: The pharmaceutical companies promoting orphan drugs are<br />

exonerated from the taxes <strong>and</strong> contributions pharmaceutical companies<br />

owe to the Sickness Insurance <strong>and</strong> the Afssaps. 59<br />

• 2002: A special funding <strong>for</strong> commercialised orphan drugs is integrated in<br />

the hospitals’ budget <strong>for</strong> innovative drugs. 59<br />

• 2006: Recognition of the “Fédération des Maladies Orphelines” dd as the<br />

only publicly recognised representative ee .<br />

Measures taken to increase the knowledge about rare diseases:<br />

• 2000-2005: the network ‘Genhomme’ was established to provide an<br />

answer to the scientific <strong>and</strong> economical challenges of the human<br />

genomics.<br />

• 2001: establishment of the “Plate<strong>for</strong>me Maladies <strong>Rare</strong>s” grouping all<br />

actors devoted to patients with rare diseases.<br />

• 2001: establishment of the “Comité de Génétique Clinique” to support<br />

research <strong>and</strong> care treatment in the field of genetics.<br />

bb This chapter has partly been reviewed by Ms Annie Lorence of the Afssaps.<br />

cc Groupe d’intérêt scientifique<br />

dd http://www.maladies-orphelines.fr/<br />

ee In French: “reconnue d’utilité publique”


46 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

A French National Plan <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong> with ten strategic priorities was published in<br />

2004. The aim is to assure equal access to diagnostic, treatment <strong>and</strong> care taking of<br />

persons suffering of a rare disease through implementation of ten strategic priorities. A<br />

new draft is being prepared at the moment.<br />

One of the strategic priorities is to enhance care management through the<br />

establishment of centres of reference. 60 Mid-February there were 131 centres of<br />

reference who were awarded the label by the Health Minister <strong>for</strong> five years. The<br />

centres have a double role: they are an expert centre <strong>for</strong> 1 or more diseases <strong>and</strong> they<br />

are a resource centre <strong>for</strong> patients coming from outside the region. A second type of<br />

centres are the qualified centres whose aim is to assume responsibility <strong>for</strong> treatment<br />

<strong>and</strong> follow-up of the patient close to their home, <strong>and</strong> to participate in the entirety of<br />

the centres of references’ tasks. These qualified centres take in charge patients that can<br />

not be treated in a reference centre. 61<br />

There are several incentives to stimulate orphan drug development: 62<br />

• Research support through national funding programmes: GIS-<strong>Rare</strong><br />

diseases, Hospital Programme of Clinical Research (Programme Hospitalier<br />

de Recherche Clinique);<br />

• During development: Free scientific advice of Afssaps;<br />

• Budgetary incentives: tax exemption of the Sickness Insurance <strong>and</strong> the<br />

Afssaps.<br />

4.4.2 Marketing Authorisation<br />

<strong>Orphan</strong> drugs obtain Marketing Authorisation through the centralised procedure at<br />

EMEA (see previous chapter).<br />

4.4.3 Reimbursement<br />

After having obtained a Marketing Authorisation of EMEA, the MAH will introduce a<br />

dem<strong>and</strong> <strong>for</strong> reimbursement at the HAS.<br />

Figure 4.9 : Introduction process of orphan drug in France: 2 steps<br />

Source: Meyer F. <strong>Orphan</strong> <strong>Drugs</strong>: How Are They Assessed / Appraised in France? In: HAS (ed). 25<br />

February 2008, p.4.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 47<br />

The dem<strong>and</strong> <strong>for</strong> inclusion on the reimbursement list is examined by the Transparency<br />

Committee of the HAS. The Transparency Committee renders an advice on the clinical<br />

added value (SMR: Service Médical Rendu) <strong>and</strong> the improvement in the clinical added<br />

value (ASMR: Amélioration du Service Médical Rendu) as compared with existing<br />

therapies. The Committee then proposes a positive or negative advice to the Health<br />

<strong>and</strong> Social Security Ministers relating to the reimbursement of the drug.<br />

There are two criteria <strong>for</strong> inclusion in the reimbursement list: 63<br />

1. Clinical added value: takes into account the indication (disease characteristics<br />

<strong>and</strong> severity) <strong>and</strong> the drug characteristics (clinical effectiveness <strong>and</strong> impact on<br />

public health). If the added value is insufficient, no reimbursement takes place.<br />

2. The improvement of clinical added value: this is the clinical improvement<br />

compared to existing therapies. There are five levels<br />

o ASMR Level I, II <strong>and</strong> III: innovative drugs (recognized added value) are<br />

eligible <strong>for</strong> faster access at a better price;<br />

o ASMR Level IV: minor improvement – product eligible <strong>for</strong> a higher<br />

price than comparators;<br />

o ASMR Level V: no improvement – reimbursement possible if costs are<br />

inferior to comparators.<br />

The Ministry of Health decides on the reimbursement of the drug.<br />

In figure 4.10, the first table provides an overview is given of the SMR since 2002 <strong>and</strong> of<br />

the ASMR <strong>for</strong> 2007. Since 2002, 35 orphan drugs have received a favourable clinical<br />

added value. The second table shows that orphan drugs score better on the ASRM level<br />

than non-orphan drugs.<br />

Figure 4.10 : Overview of the assessment of orphan drugs in France<br />

SMR (since 2002)<br />

Year N<br />

2002 1<br />

2003 6<br />

2004 5<br />

2005 3<br />

2006 6<br />

2007 12<br />

Total 35<br />

Level ASMR 2007 <strong>for</strong> orphan <strong>and</strong> nonorphan<br />

drugs<br />

N % OD % all drugs<br />

ASMR I Major 3 10% 1%<br />

ASMR II Important 13 43% 4%<br />

ASMR III Moderate 8 27% 6%<br />

ASMR IV Minor 4 13% 5%<br />

ASMR V No improvement 2<br />

30<br />

7% 84%<br />

Source: Meyer F. <strong>Orphan</strong> <strong>Drugs</strong>: How Are They Assessed / Appraised in France? In: HAS (ed). 25<br />

February 2008, p.12.<br />

The Transparency Committee also: 64<br />

• Makes an estimate of the target population;<br />

• Gives advice to the prescribers on the drug’s place within therapy;<br />

• Determines the limits of the currently available data <strong>and</strong> request<br />

additional data.


48 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Another actor intervening in the reimbursement decision is the National Health<br />

Insurance Fund. It<br />

• fixes reimbursement rates <strong>for</strong> drugs within the conditions <strong>and</strong> limits fixed<br />

by the State;<br />

• classifies drugs into categories on the basis of the National Health<br />

Authority assessment of the clinical added value;<br />

• decides which acts <strong>and</strong> per<strong>for</strong>mances will be reimbursed. 65<br />

4.4.3.1 Compassionate use<br />

<strong>Orphan</strong> drugs can be delivered to patients without having first received a Marketing<br />

Authorisation, through clinical trials, authorisation <strong>for</strong> temporary usage (ATU) <strong>and</strong><br />

hospital preparations. Experimental drugs can be administrated in clinical trials <strong>and</strong> to<br />

hospital preparations <strong>for</strong> which there is no pharmaceutical speciality available or<br />

adapted. Furthermore, innovative drugs may receive an ATU of the Afssaps if there is a<br />

public health need. The drug must fulfil several criteria: it is a treatment <strong>for</strong> a serious or<br />

rare disease; no therapeutic alternative is available; it has a positive risk/benefit <strong>and</strong> it is<br />

<strong>for</strong> temporary use. The evaluation will take into account aspect of the drug (quality,<br />

security <strong>and</strong> efficacy) <strong>and</strong> the medical environment (disease <strong>and</strong> alternatives). Examples<br />

or medicinal products that use the ATU are: Thalidomide®, Aldurazyme®, Cerezyme®,<br />

Fabrazyme® <strong>and</strong> Carbaglu®. 66<br />

4.4.3.2 Off-label use<br />

4.4.4 Pricing<br />

Off-label use of an orphan or non-orphan drug is possible <strong>for</strong> a rare disease (as defined<br />

by the European Regulation 141/2000 5 ) if the medicinal product is listed in an advice or<br />

recommendation relating to a category of sick persons of the HAS (Article L162-17-2-1<br />

of the Social Security Legal Code).<br />

The treatment <strong>and</strong> reimbursement are decided by decree of the Ministers of Health <strong>and</strong><br />

Social Security <strong>and</strong> following advice of the National Union of the Sickness Funds. The<br />

specialities, products or services being the subject of the decree can be dealt with only<br />

if their use is essential to the improvement of the health of the patient or to avoid its<br />

deterioration. They must moreover be registered explicitly in the protocol of care.<br />

The Economic Committee <strong>for</strong> Health Products of the Ministry of Health negotiates the<br />

price of an orphan drug with the pharmaceutical company in order to reach an<br />

agreement on the price-volume. 67<br />

Price setting <strong>for</strong> an orphan drug takes into account: 63<br />

• the improvement in clinical added value of the medicine;<br />

• the prices of medicines serving the same therapeutic purpose;<br />

• <strong>for</strong>ecasted or recorded sales volumes;<br />

• <strong>for</strong>eseeable <strong>and</strong> actual conditions of use of medicine;<br />

• the National Health Authority assessment;<br />

• reference prices in Irel<strong>and</strong>, Italy, Portugal, Spain <strong>and</strong> the EU. 68<br />

4.4.5 Distribution<br />

<strong>Orphan</strong> drugs are distributed through the community pharmacies or the hospital<br />

pharmacies. For one third of the orphan drugs prescription by <strong>and</strong> delivery through the<br />

hospital pharmacy <strong>for</strong> hospitalised patients is obligatory.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 49<br />

4.4.6 Prescribing<br />

The orphan drug will be reimbursed if the rare disease is one of the indications.<br />

Otherwise, the drug can still be prescribed, but it is not reimbursable. 59<br />

The prescription must be delivered by a MD specialist either working on its own<br />

(minority of orphan drugs) or within a hospital (majority of orphan drugs). But the first<br />

prescription has to be delivered by a centre of reference (if such centre exists <strong>for</strong> the<br />

disease at issue).<br />

It is the Social Security that is in charge of reimbursement.<br />

Key points<br />

• France has in place specific centres of reference, policy measures, <strong>and</strong><br />

research incentives on rare diseases/orphan drugs.<br />

• <strong>Orphan</strong> drugs are registered through the EMEA centralised procedure.<br />

Specific legislation governs compassionate use of orphan drugs.<br />

• <strong>Orphan</strong> drug prices are fixed by the Economic Committee <strong>for</strong> Health<br />

Products of the Ministry of Health.<br />

• The reimbursement procedure considers the clinical added value. <strong>Orphan</strong><br />

drugs are fully or partially reimbursed by social insurance.<br />

• For some orphan drugs, prescription <strong>and</strong> delivery through hospital<br />

pharmacies is compulsory.<br />

4.5 ITALY ff<br />

4.5.1 Institutional context<br />

The Italian Medicines Agency (AIFA - Agenzia Italiana del Farmaco) is in charge of the<br />

introduction of orphan drugs on the Italian medicine market. The AIFA has also set up a<br />

fund of around 45 millions Euro a year, of which half is used to the reimbursement of<br />

orphan <strong>and</strong> ‘life saving’ drugs <strong>and</strong> the other half is aimed at supporting independent<br />

research, drug in<strong>for</strong>mation programs <strong>and</strong> pharmacovigilance. 69 This funding program<br />

<strong>for</strong> independent clinical research on drugs is open to researchers working in public <strong>and</strong><br />

non profit institutions. One of the research areas of the program is dedicated to orphan<br />

drugs <strong>for</strong> rare diseases. At the start of 2009, three calls <strong>for</strong> proposals (2005-2007) have<br />

been concluded <strong>and</strong> 69 studies have received funding in the area of rare diseases.<br />

Several incentives <strong>for</strong> promoting non-profit research were issued in an ad hoc<br />

regulation of 2004:<br />

• the fees of the ethics committee are waived;<br />

• the National Health Service (NHS) can reimburse the study drugs;<br />

• <strong>and</strong> patients’ insurance costs are financed by the study institution. 69<br />

In every Italian region there are one or more Regional Centres, which act as reference<br />

centres in the region <strong>and</strong> are authorised to diagnose rare diseases <strong>and</strong> to prescribe<br />

orphan medicines. There is also a National Centre <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong> at the National<br />

Institute of Health, which coordinates the activity of the regional centres, carries out<br />

scientific research <strong>and</strong> public health activities, including cooperation with patient<br />

associations. 70<br />

Three National Healthcare Plans (1998-2000; 2003-2005; 2006-2008) <strong>and</strong> Regional<br />

Health Plans were <strong>for</strong>mulated where rare diseases were addressed. The first National<br />

Plan has defined rare diseases as a priority <strong>for</strong> public health.<br />

ff This chapter has been reviewed by Dr. Pietro Folino Gallo of the Italian Medicines Agency.


50 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

A National Network <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong> <strong>and</strong> a National Registry of <strong>Rare</strong> <strong>Diseases</strong> were<br />

established in 2001. 60 The National Network is composed of a network of hospitals <strong>and</strong><br />

referral centres organised by region where patients can be diagnosed <strong>and</strong> treated <strong>for</strong><br />

free <strong>for</strong> about 500 rare diseases. The aims 71 of the Network are:<br />

• Prevention: implementation of prevention activities,<br />

• Surveillance: develop epidemiological surveillance,<br />

• Diagnosis: implement both diagnosis <strong>and</strong> care intervention,<br />

• Treatment: improve health operators’ training,<br />

• Promote citizens in<strong>for</strong>mation.<br />

The National Registry of <strong>Rare</strong> <strong>Diseases</strong> is to be completed by regional centres. The<br />

registry’s general objectives are national <strong>and</strong> regional health planning <strong>and</strong> surveillance of<br />

rare diseases. The specific objectives are the estimation of incidence <strong>and</strong>/or prevalence,<br />

the definition of st<strong>and</strong>ardized diagnostic <strong>and</strong> therapeutic protocols, <strong>and</strong> the<br />

improvement of the collaboration among health care operators.<br />

4.5.2 Marketing Authorisation<br />

<strong>Orphan</strong> drugs obtain Marketing Authorisation through the centralised procedure at<br />

EMEA (see previous chapter).<br />

4.5.2.1 Compassionate use<br />

Compassionate use of orphan drugs waiting <strong>for</strong> approval is possible <strong>and</strong> financed<br />

through a special fund called “Fondo AIFA 5%”. The aim of this fund is threefold: to<br />

improve knowledge on efficacy <strong>and</strong> safety of orphan drugs; to improve knowledge on<br />

efficacy <strong>and</strong> safety of non-licensed / non-marketed orphan drugs <strong>and</strong> to promote access<br />

to orphan drugs waiting <strong>for</strong> a Marketing Authorisation. 72<br />

This not only applies to orphan drugs, but also to trials on rare diseases.<br />

In 2008, four orphan molecules were reimbursed by the NHS through the Fondo AIFA<br />

5%.<br />

4.5.2.2 Off-label procedure<br />

Italy also knows an off-label procedure regulated by Law 648/96. The Technical<br />

Committee of the AIFA can include a given medication into a special list allowing it to<br />

be prescribed at NHS (National Health Service) charge, if <strong>for</strong> a specific disease there is<br />

no therapeutic alternative. There are three types of medical products that can be<br />

included:<br />

• innovative drugs whose sale is authorised abroad, but not in Italy;<br />

• drugs not yet authorised but which underwent clinical trials;<br />

• <strong>and</strong> drugs to be used <strong>for</strong> a therapeutic indication other than the one<br />

which has been authorised.<br />

At present fourteen orphan molecules are reimbursed <strong>for</strong> rare diseases in off-label use<br />

by the NHS. 73<br />

4.5.3 Reimbursement<br />

<strong>Orphan</strong> drugs that have obtained an EMEA Marketing Authorisation can apply <strong>for</strong><br />

reimbursement in Italy.<br />

In order to be reimbursed, any medical products, including orphan drugs, has to meet a<br />

number of criteria:<br />

• it covers an unmet need being a relevant disease without any efficient<br />

therapy;<br />

• existing therapies <strong>for</strong> a relevant disease are not satisfactory;<br />

• the product has a better benefit-risk ratio than existing therapies;<br />

• it has a socio-economic benefit. 72


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 51<br />

These criteria are evaluated at the Scientific-Technical Committee (Commissione<br />

Tecnico-Scientifica) <strong>and</strong> at the Pricing <strong>and</strong> Reimbursement Committee. Both these<br />

Committees are consultative bodies within the Italian Medicines Agency.<br />

Because, by definition, an orphan drug covers an unmet need, most of the orphan drugs<br />

are at the charge of the Italian NHS.<br />

Figure 4.11 : Availability of licensed orphan drugs in Italy (October 2007)<br />

70<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

4.5.4 Pricing<br />

0<br />

68<br />

21<br />

Reimbursed <strong>and</strong><br />

marketed<br />

6<br />

2<br />

Reimbursed, but not<br />

marketed<br />

% Number of molecules<br />

26<br />

8<br />

Not available<br />

Source: Folino Gallo P. <strong>Orphan</strong> <strong>Drugs</strong> in Italy. Accommodating orphan drugs: balancing innovation<br />

<strong>and</strong> financial stability. Accommodating orphan drugs: balancing innovation <strong>and</strong> financial stability; 25<br />

February 2008; London.<br />

Two committees of the Italian Medicines Agency are involved in the pricing <strong>and</strong><br />

reimbursement procedure of medical products. The Scientific-Technical Committee<br />

takes decisions if a medical product can be reimbursed <strong>and</strong> positive list revisions, while<br />

the Pricing <strong>and</strong> Reimbursement Committee assesses the applications <strong>and</strong> negotiates<br />

with the MAHs according to the m<strong>and</strong>ate received by the Scientific-Technical<br />

Committee.<br />

The procedure <strong>for</strong> pricing pharmaceutical products reimbursed by the NHS is fixed by<br />

law. First, the Scientific-Technical Committee decides whether or not a product can be<br />

reimbursed. If yes, a price negotiation takes place between the Pricing <strong>and</strong><br />

Reimbursement Committee <strong>and</strong> the MAH. If the negotiation turns out positively, the<br />

product will be reimbursed.<br />

The product will then be listed in one of three drug classes: class A includes essential<br />

products <strong>and</strong> products <strong>for</strong> chronic diseases that are 100% reimbursed by the NHS; class<br />

H includes products that are 100% reimbursed through the hospital; <strong>and</strong> class C<br />

includes all other products which are not reimbursed because the health authorities<br />

intend to discourage their use (these products have a low evidence level <strong>and</strong>/or a low<br />

benefit / risk ratio).<br />

Medicines included in class C are <strong>for</strong> example antiobesity agents <strong>and</strong> benzodiazepines.<br />

Products <strong>for</strong> minor ailments are not reimbursed.<br />

Products of class C can still be used <strong>for</strong> free at a hospital level.


52 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Price determination criteria are: 68<br />

• the efficacy of the product in relation to existing therapies, taking into<br />

account its degree of innovation, the clinical relevance, incidence <strong>and</strong><br />

prevalence of the disease it intends to threat, possible reductions in<br />

hospitalisation, <strong>and</strong> quality of life improvements;<br />

• price comparisons with other countries (but a <strong>for</strong>mal external price<br />

reference system was withdrawn in 2004);<br />

• <strong>for</strong>ecasts of sales, including revenues derived from licensing agreements;<br />

• financial factors, related investment, spill over effects on employment, <strong>and</strong><br />

exports.<br />

Prices are generally revised after two years, but both AIFA <strong>and</strong> company can at any<br />

moment request a revision of the contract. The company can even lower the price<br />

without permission, <strong>for</strong> instance after patent expiration <strong>and</strong> the introduction of a<br />

generic competitor, but a notification of the new price is needed.<br />

4.5.5 Distribution<br />

<strong>Orphan</strong> drugs are distributed through hospital <strong>and</strong> community pharmacies, <strong>and</strong> by<br />

health authorities.<br />

4.5.6 Prescribing<br />

The orphan drug is prescribed by a specialist MD member of a centre of reference.<br />

A control mechanism exists <strong>for</strong> some rare diseases under the <strong>for</strong>m of national registers<br />

(which are not much populated). AIFA also has a tracking system (traceability) <strong>for</strong><br />

monitoring the movement of every pack (including orphan drugs) from the<br />

manufacturer via the wholesale to the hospitals <strong>and</strong> pharmacies. Some regions have<br />

local systems to match the prescriber/dispenser <strong>and</strong> the patient.<br />

Figure 4.12 : <strong>Orphan</strong> <strong>Drugs</strong> subjected to registration<br />

Aldurazyme® <strong>Orphan</strong> Drug Register<br />

Cabarglu® <strong>Orphan</strong> Drug Register<br />

Myozyme® <strong>Orphan</strong> Drug Register<br />

Somavert® <strong>Orphan</strong> Drug Register<br />

Ventavis® <strong>Orphan</strong> Drug Register<br />

Zavesca® <strong>Orphan</strong> Drug Register<br />

Nexavar® Oncologic Register<br />

Xagrid® Oncologic Register<br />

Sutent® Oncologic Register<br />

Source: Folino Gallo P. <strong>Orphan</strong> <strong>Drugs</strong> in Italy. Accommodating orphan drugs: balancing innovation<br />

<strong>and</strong> financial stability. Accommodating orphan drugs: balancing innovation <strong>and</strong> financial stability; 25<br />

February 2008; London.<br />

Conditions may be applied to the prescription of orphan drugs. One of these conditions<br />

is the registration of the treatment into a national register (especially <strong>for</strong> cancers). This<br />

means that the hospital doctor must list the patient into the register <strong>and</strong> complete the<br />

appropriate <strong>for</strong>m (registration, treatment start, follow-up, …). Hospital pharmacists can<br />

dispense the orphan drugs only upon a written request with attached the register<br />

sheet. 69<br />

In some regions a dispensation fee of around 2 Euro is imposed, but patients with a rare<br />

disease are generally exempted from this fee.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 53<br />

Key points<br />

• Italy has in place specific centres of reference, policy measures, <strong>and</strong> research<br />

incentives on rare diseases/orphan drugs.<br />

• <strong>Orphan</strong> drugs are registered through the EMEA centralised procedure only.<br />

Specific legislation governs compassionate use <strong>and</strong> off-label use of orphan<br />

drugs.<br />

• <strong>Orphan</strong> drug prices are fixed by the Pricing <strong>and</strong> Reimbursement<br />

Committee.<br />

• The reimbursement procedure considers budget impact <strong>and</strong> costeffectiveness.<br />

<strong>Orphan</strong> drugs are fully reimbursed by the National Health<br />

Service.<br />

• <strong>Orphan</strong> drugs are distributed through hospital <strong>and</strong> community pharmacies,<br />

<strong>and</strong> by health authorities.<br />

• The orphan drug is prescribed by a specialist MD member of a centre of<br />

reference. Conditions, such as registration of the treatment in a national<br />

register, are applied to the prescription of orphan drugs.<br />

4.6 THE NETHERLANDS gg<br />

4.6.1 Institutional context<br />

The applicable rare diseases definition in the Netherl<strong>and</strong>s is the EU definition.<br />

The institution in charge of rare diseases is the Ministry of Health.<br />

There are no official centres of rare diseases, but the eight university medical centres<br />

fulfil the role of main clinical expertise centres <strong>for</strong> specific rare diseases <strong>and</strong> can function<br />

as centres of specific rare diseases. For some rare diseases also other (top clinical)<br />

hospitals may function as centres <strong>for</strong> rare diseases.<br />

Several policy measures were taken in order to promote the development of orphan<br />

drugs: 74<br />

• A Steering Committee <strong>Orphan</strong> <strong>Drugs</strong> was established in 2001 in order<br />

“to encourage the development of orphan drugs <strong>and</strong> to improve the<br />

situation of patients with a rare disease, especially to strengthen the<br />

transfer of in<strong>for</strong>mation on rare diseases”. hh The Committee is also a<br />

member <strong>and</strong> work package leader of the European project European<br />

(European Project <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong> National Plans Development, 2008-<br />

2011) that is preparing recommendations <strong>for</strong> the Member States on how<br />

to write a national plan on rare diseases <strong>and</strong> orphan drugs.<br />

• An orphan product developer was appointed in 2006 within the Dutch<br />

Organisation <strong>for</strong> Health Research <strong>and</strong> Development (ZonMw) ii to in<strong>for</strong>m<br />

academia <strong>and</strong> enterprises (especially SME’s) about the European<br />

Regulation on <strong>Orphan</strong> Medicinal Products in an active way (by means of<br />

visits, seminars, articles, etc.). This person (R. de Rue) has been appointed<br />

<strong>for</strong> four years. After that it will be examined if the function can be h<strong>and</strong>ed<br />

on to the Medicines Evaluation Board or to an industry plat<strong>for</strong>m.<br />

gg This chapter has been reviewed by Dr. Sonja van Weely of the Dutch Steering Committee <strong>Orphan</strong> <strong>Drugs</strong><br />

<strong>and</strong> Dr Gepke Delwel of the Health Care Insurance Board<br />

hh www.weesgeneesmiddelen.nl<br />

ii www.zonmw.nl


54 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

• A PhD student (H. Hoekstra) was appointed in 2005 in order to study the<br />

factors of success <strong>and</strong> failure in orphan drug development <strong>and</strong> this in close<br />

collaboration with the Steering Committee <strong>and</strong> the orphan drug<br />

developer. 75<br />

• The Dutch registration fee <strong>for</strong> a medicinal product can be waived if the<br />

medicinal product is already registered in one or several other EU<br />

Member States <strong>and</strong> the prevalence of the indicated disease is less than 1<br />

in 200,000 inhabitants in the Netherl<strong>and</strong>s.<br />

• In 2007 a new research programme <strong>for</strong> rare diseases <strong>and</strong> orphan drugs<br />

was developed in order to develop therapies <strong>for</strong> rare diseases, but no<br />

<strong>for</strong>mal decision of the Ministry of Health on the funding of this<br />

programme has been taken until now.<br />

• An <strong>Orphan</strong> Drug Designation Support Programme was launched in<br />

January 2009: Dutch enterprises can apply <strong>for</strong> a grant to compensate the<br />

application costs <strong>for</strong> the EMEA <strong>Orphan</strong> Drug Designation. jj<br />

• In April 2009 the Dutch <strong>Orphan</strong> Registry Consortium was launched, a<br />

multidisciplinary group that will use best practices to build a registry<br />

frame work <strong>for</strong> inborn errors of metabolism.<br />

The Netherl<strong>and</strong>s (represented by Zoom <strong>and</strong> the Steering Committee) are partner in E-<br />

<strong>Rare</strong> (ERA-Net <strong>for</strong> research programmes on rare diseases) 69 , a research network<br />

funded by the European Commission, providing a setting to bring together clinicians <strong>and</strong><br />

scientists <strong>and</strong> gather research infrastructure, patient cohorts <strong>and</strong> related biological<br />

material on a European scale. Zoom is the leader of work package 5 that focuses on the<br />

opening of programmes to encourage a multidisciplinary approach.<br />

Zoom provides funding through several research programmes <strong>for</strong> research on rare<br />

diseases, e.g.<br />

• The Innovative Research Incentives Scheme;<br />

• The Gene Therapy subsidy scheme.<br />

In<strong>for</strong>mation on rare diseases <strong>and</strong> orphan drugs is disseminated by different actors:<br />

• Royal Dutch Society <strong>for</strong> Pharmacists (KNMP) created the website<br />

www.farmanco.knmp.nl where in<strong>for</strong>mation on European registered<br />

orphan drugs can be found with in<strong>for</strong>mation on their reimbursement in<br />

the Netherl<strong>and</strong>s;<br />

• Patient organisations <strong>for</strong> (a group of) rare diseases (they can obtain<br />

funding indirectly from the Ministry of Health).<br />

4.6.2 Marketing Authorisation<br />

There is no national procedure of Marketing Authorisation <strong>for</strong> orphan drugs. All orphan<br />

drugs have to be registered at the EMEA, but the Medicines Evaluation Board (MEB) is<br />

involved in evaluating orphan drugs <strong>for</strong> the EMEA. As an independent medicinal<br />

products knowledge centre, the MEB evaluates the balance efficiency-safety (this is<br />

efficiency, risks, quality) of drugs <strong>for</strong> humans <strong>and</strong> animals, thus the advantages versus<br />

disadvantages. 76 The MEB’s report contains product in<strong>for</strong>mation <strong>for</strong> MD <strong>and</strong> pharmacist<br />

<strong>and</strong> an instruction leaflet <strong>for</strong> the patient. The MEB will continue to follow-up<br />

in<strong>for</strong>mation on the drug even after it entered the market.<br />

jj http://www.zonmw.nl/nl/subsidie/subsidiekalender/subsidieronde/item/subsidieregeling-orph<strong>and</strong>esignation-dossier-odd-support/


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 55<br />

Compassionate use<br />

There is no specific policy <strong>for</strong> orphan drugs, but there is a general policy <strong>for</strong><br />

compassionate use. In exceptional cases, compassionate use is possible if: 77<br />

• There is a MD declaration (named patient);<br />

• It concerns a serious condition <strong>for</strong> which no alternative drug is on the<br />

market <strong>and</strong> of which the drug is awaiting a Marketing Authorisation. In<br />

that case, the MAH can apply <strong>for</strong> ‘the compassionate use programme’.<br />

Off-label use<br />

The off-label procedure is the same <strong>for</strong> orphan drugs <strong>and</strong> non-orphan drugs. Off-label<br />

use is accepted if scientific evidence attests of an added value of the treatment with the<br />

drug, the drug is rational <strong>and</strong> justified, but which has not (yet) been evaluated by the<br />

MEB. 78 The patient must be in<strong>for</strong>med of the off-label treatment. 78<br />

4.6.3 Reimbursement<br />

Extramural drugs, i.e. drugs that are prescribed by General Practioners <strong>and</strong> are used in<br />

the out-patient setting, can apply <strong>for</strong> reimbursement after the registration. All<br />

manufacturers do apply <strong>for</strong> reimbursement at the Ministry of Health. Consequently, the<br />

Health Care Insurance Board (HCIB) will per<strong>for</strong>m the assessment <strong>and</strong> appraisal<br />

procedure <strong>for</strong> the drug based on the submitted dossier by the manufacturer. Most<br />

drugs are reimbursed; co-payments are possible but are rare in the Netherl<strong>and</strong>s.<br />

Intramural drugs or hospital-based drugs i.e. drugs that are prescribed by medical<br />

specialists within hospitals are paid by the hospital budget. All medical products that are<br />

included in the official treatment guidelines of the physicians are available to patients <strong>and</strong><br />

have to be paid <strong>for</strong> by the hospitals. Since hospital budgets are limited, it is difficult to<br />

ensure equal access of expensive drugs to all patients. To overcome this ‘postcode<br />

prescription’ by hospitals, policy measures have been issued. Through these policy<br />

measures expensive hospital-based (orphan) drugs can apply <strong>for</strong> additional funding.<br />

<strong>Orphan</strong> drugs that are listed on the policy measure will get a full funding, so the costs<br />

<strong>for</strong> these drugs are fully covered by means of those additional budgets. In case of<br />

expensive hospital based drugs a 80% funding is provided to the hospitals. Hospitals can<br />

apply <strong>for</strong> additional funding at the Dutch Care Authority (NZa). The HCIB will<br />

subsequently assess <strong>and</strong> appraise the request based on the submitted dossier by the<br />

applicant (hospitals, physicians <strong>and</strong> manufacturer are involved). The additional funding is<br />

always conditional, i.e. temporally, additional in<strong>for</strong>mation needs to be collected through<br />

outcomes research. After three years a reassessment takes place in order to assess<br />

whether listing / funding will continue.<br />

The assessment <strong>and</strong> appraisal procedures <strong>for</strong> extramural <strong>and</strong> intramural drugs are<br />

different: reimbursement versus additional funding <strong>and</strong> assessment versus a two tiered<br />

process based on coverage with evidence. The assessment criteria are not very<br />

different. For extramural drugs the HCIB assess the therapeutic value of the drug in<br />

comparison with the existing st<strong>and</strong>ard treatment - assessment of the place of the new<br />

drug in the therapy; the cost-effectiveness of the drug <strong>and</strong> the budget impact of the drug<br />

<strong>for</strong> the pharmacy budget. For intramural drugs, the assessment criteria <strong>for</strong> temporally<br />

listing are the therapeutic value, the cost prognosis, the cost-effectiveness indication<br />

<strong>and</strong> the proposal <strong>for</strong> outcomes research. After three years the reassessment criteria<br />

are: the therapeutic value; the actual costs of the medical product; the costeffectiveness<br />

<strong>and</strong> the efficient prescription. The efficient prescription of the drug in<br />

Dutch hospitals is based on data collected through outcomes research in the Dutch<br />

clinical practice. Also the cost-effectiveness will be based on those data, in addition to<br />

other data sources like the clinical registration trials.


56 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

4.6.3.1 Extramural treatment<br />

In order to be included in the Medicine Reimbursement System (GVS), by which<br />

extramural drugs are reimbursed, the manufacturer must <strong>for</strong>mally request a submission<br />

<strong>for</strong> reimbursement at the Ministry of Health. The HCIB will subsequently per<strong>for</strong>m the<br />

assessment <strong>and</strong> appraisal based on the submitted dossier by the manufacturer. The<br />

director of the HCIB (or the Board of the HCIB) will give a motivated advice to the<br />

Minister of Health regarding reimbursement of the drug in the reimbursement system<br />

<strong>and</strong> on what list the drug should be placed. The reimbursement system contains a<br />

positive list of all reimbursed drugs. List 1A consists of groups of medical products that<br />

are interchangeable, <strong>for</strong> each group a reimbursement limit exists (reference price<br />

system). List 1B consists of unique medical products; no reimbursement limit exists <strong>for</strong><br />

those medical products although prices are restricted due to the Act on Medicine<br />

prices (the price of the drug in neighbouring countries is taken as a reference). Both <strong>for</strong><br />

drugs listed on list 1A <strong>and</strong> <strong>for</strong> those listed on list 1B special conditions <strong>for</strong><br />

reimbursement may exist; these are listed on list 2. For example the drug is only<br />

reimbursed <strong>for</strong> a small group of patients, or must be prescribed by a specialist.<br />

Next to the reimbursement conditions on List 2 health care insurance companies may<br />

also give restrictions , e.g. that the medicine may only be prescribed by a specialist or a<br />

specific prescriber (e.g. a specialist with experience in a particular disease).<br />

Based on the EU transparency regulation, the assessment <strong>and</strong> appraisal procedure<br />

followed by the decision of the Minister of Health may last 90 days. In practice, these<br />

time lines are in general met <strong>for</strong> drugs listed on list 1A, the ones listed on list 1B may<br />

take longer especially when special conditions (list 32) are involved. An ongoing<br />

assessment procedure can be suspended <strong>for</strong> three months on request of the MAH in<br />

case time is needed to collect necessary in<strong>for</strong>mation <strong>for</strong> the assessment of the drug.<br />

The procedure is the same <strong>for</strong> orphan <strong>and</strong> non-orphan drugs. The evaluation procedure<br />

is done by the Committee <strong>for</strong> Pharmaceutical Aid (CPA) of the HCIB <strong>and</strong> is composed<br />

of three parts corresponding to the three components of the application file: the<br />

pharmacotherapeutic (therapeutic value) evidence, the pharmaco-economic evaluation<br />

(cost-effectiveness) <strong>and</strong> the budget impact provided by the MAH. The CPA will first<br />

assess if the drug is interchangeable or not (this means that an alternative is available). If<br />

yes, the drug will be included on List 1A. If not interchangeable, the CPA judges the<br />

therapeutically added value <strong>and</strong> the cost-effectiveness of the product. It will there<strong>for</strong>e<br />

look at the added therapeutic value (assessing the pharmatherapeutic evidence), the<br />

cost-effectiveness (assessing the pharmaco-economic evaluation) <strong>and</strong> the budget<br />

impact kk 79 80<br />

. If these criteria are met, the product can be included on List 1B.<br />

If the indication is a rare disease <strong>and</strong> no alternative treatment as is the case <strong>for</strong> most<br />

orphan drugs, the MAH may ask <strong>for</strong> dispensation of the pharmaco-economic evaluation.<br />

This request is judged by the HCIB <strong>and</strong> is frequently given. A dispensation may also be<br />

asked <strong>for</strong> drugs that have a low budget impact (€500,000 annually).<br />

kk The budget impact takes into account (among other things): the number of patients; possible new<br />

alternatives; the drug’s market share; the possible off-label use; <strong>and</strong> applicable costs.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 57<br />

4.6.3.2 Intramural treatment<br />

Due to limiting hospital budgets equal access to medicines became hampered, especially<br />

<strong>for</strong> the treatment of certain cancers. To overcome the postcode prescription additional<br />

funding is provided through the following policy measures: ll<br />

• Policy measure “Expensive drugs” <strong>for</strong> hospitals: 80% reimbursement if<br />

purchase costs of a specific mm orphan drug account <strong>for</strong> more than 0.5% of<br />

the total drug cost of all hospitals on a macro level.<br />

• Policy measure “<strong>Orphan</strong> drugs” <strong>for</strong> academic hospitals: if the orphan drug<br />

costs account <strong>for</strong> more than 5% of the hospital’s drug budget, the surplus<br />

will be fully reimbursed to the hospital.<br />

These policy measures provide provisional funding <strong>for</strong> three years (temporary listing)<br />

<strong>and</strong> require collection of more evidence on the clinical <strong>and</strong> cost effectiveness of drugs<br />

fitting in either group through outcomes research. After maximum three years, the<br />

HCIB re-appraises the evidence that has been developed as a result of the additional<br />

studies, <strong>and</strong> on this basis it reviews its decisions on the product listing. When the<br />

evidence meets the expectations the drug will be kept on the list (definitive listing). This<br />

research fosters the development of expertise <strong>for</strong> specific rare diseases: a special<br />

research programme at ZonMw has been dedicated to fund research on effectiveness of<br />

expensive (innovative) drugs <strong>and</strong> expensive orphan drugs <strong>and</strong> research on development<br />

of methodology <strong>for</strong> Health Technology Assessments. Funding of the first projects in this<br />

research programme has started in 2008, so there are no results as yet. 74<br />

In 2005, a new instrument <strong>for</strong> per<strong>for</strong>mance-oriented costing system <strong>for</strong> hospital care<br />

<strong>and</strong> <strong>for</strong> medical mental health care was introduced <strong>for</strong> hospital treatments, the<br />

Diagnosis <strong>and</strong> Treatment Combinations (DTC). A DTC includes all the activities <strong>and</strong><br />

actions per<strong>for</strong>med by the hospital <strong>and</strong> medical specialist in response to the patient’s<br />

need <strong>for</strong> care. Within a DTC, hospital output prices are determined based on actual<br />

production costs. The DTC costs are reimbursed by the patient’s insurance company.<br />

For rare diseases there are not many diagnosis <strong>and</strong> treatment combinations <strong>and</strong><br />

there<strong>for</strong>e, in many cases, hospitals are responsible <strong>for</strong> identifying <strong>and</strong> funding (from<br />

their budget) the treatments outside the DTC provided to patients. 74<br />

4.6.3.3 Current situation<br />

Of the 47 orphan drugs having received a Marketing Authorisation of the EMEA:<br />

• Eight orphan drugs are financed under the “<strong>Orphan</strong> <strong>Drugs</strong>” policy rule<br />

<strong>and</strong> one drug has applied <strong>for</strong> this;<br />

• One orphan drug is financed under the “Expensive drugs” policy rule;<br />

• Five orphan drugs are included on List 1A (reimbursement with<br />

conditions);<br />

• Eighteen orphan drugs are included on List 1B (100% reimbursement);<br />

• Eight orphan drugs are available, but not reimbursed;<br />

• Three orphan drugs still have to apply <strong>for</strong> reimbursement or are ongoing.<br />

This means that of the 47 EMEA orphan drugs, only three are not available in the<br />

Netherl<strong>and</strong>s.<br />

ll<br />

These policy rules have been developed in order to ensure equal access of drugs to patients across<br />

hospitals in the Netherl<strong>and</strong>s.<br />

mm Most orphan drugs apply <strong>for</strong> the policy measure <strong>for</strong> orphan drugs in order to obtain full funding. It is<br />

possible to apply <strong>for</strong> the policy measure on expensive drugs.


58 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

4.6.4 Pricing<br />

There are two mechanisms to regulate the price of drugs. The price of orphan drugs is<br />

regulated in the same way as non-orphan drugs.<br />

One mechanism is the incorporation of drugs in the Medicine Reimbursement system in<br />

case of extramural treatment. Specific clusters of drugs that are assumed to be<br />

therapeutically equivalent are listed on the so-called List 1A. A maximum level of<br />

financing is established <strong>for</strong> each cluster on the List A. List 1B products are drugs used<br />

<strong>for</strong> extramural treatment that do not have a therapeutically equivalent <strong>and</strong> do not have<br />

a maximum level of price. Until now most orphan drugs that are used <strong>for</strong> extramural<br />

treatment are on List 1B. The price is in this case considered together with the<br />

reimbursement decision.<br />

The second mechanism is the Regulation on maximum prices of medicinal products that<br />

fixes the maximum price that a manufacturer can ask <strong>for</strong> a medicinal product (listed in<br />

List 1A). The average prices (ex-factory prices) of Belgium, UK, Germany <strong>and</strong> France<br />

are used to calculate the maximum price scheme. A maximum price is set <strong>for</strong> each<br />

product with a given active substance, strength <strong>and</strong> <strong>for</strong>mulation (constituting clusters of<br />

products). 74<br />

A price revision takes place every six months according to a basket of prices from<br />

Belgium, UK, Germany <strong>and</strong> France. The inclusion of UK influences the basket because of<br />

the value of the pound. This mechanism does not apply to the majority of orphan drugs<br />

as they fall under list 1B.<br />

The Regulation on maximum prices holds also <strong>for</strong> those medicinal products that are<br />

used <strong>for</strong> treatments within hospitals <strong>and</strong> that are either also used extramural treatment<br />

or are placed on the policy rule on Expensive medicines or on the policy rule of <strong>Orphan</strong><br />

drugs.<br />

4.6.5 Distribution<br />

4.6.6 Prescribing<br />

<strong>Orphan</strong> drugs are distributed through hospital <strong>and</strong> community pharmacies.<br />

The first prescription will be issued by the specialist physician or general practitioner.<br />

Different reimbursement procedures apply whether the drug is prescribed <strong>for</strong> home<br />

treatment (extramural) or is administered within the hospital setting (intramural).<br />

Home treatment costs are reimbursement by the Medicines Reimbursement System.<br />

The reimbursement level depends of whether the drugs are included on list 1A or on<br />

list 1B. <strong>Drugs</strong> listed on list 1B are 100% reimbursed, while there is a maximum<br />

reimbursement <strong>for</strong> each cluster of interchangeable products on list 1A. The patient will<br />

have to pay the surplus above the maximum reimbursement.<br />

Hospital treatment costs are (partly) taken in charge by the hospital’s budget.<br />

In case of off label use, reimbursement is automatic <strong>for</strong> a disease with prevalence less<br />

than 1:150,000 inhabitants.<br />

In some cases restrictions are imposed on the reimbursement of orphan drugs. The<br />

reimbursement conditions to be found in list 2 of the Law Care Assurance are a first<br />

type of restriction. A condition can be that the drug is only reimbursed <strong>for</strong> a specific<br />

indication, making it unavailable <strong>for</strong> off-label use. For example, miglustat can only be<br />

reimbursed if the patient has Gaucher type 1 <strong>and</strong> he or she can not be treated with<br />

imiglucerase. A second type of restriction is imposed by the health care insurance<br />

companies. For example, the medicine can only be prescribed by a specialist or a<br />

specific prescriber. 74


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 59<br />

Key points<br />

• The Netherl<strong>and</strong>s have in place policy measures <strong>and</strong> research incentives on<br />

rare diseases/orphan drugs. There are non-official centres of reference.<br />

• <strong>Orphan</strong> drugs are registered through the EMEA centralised procedure only.<br />

Specific legislation governs compassionate use <strong>and</strong> off-label use of orphan<br />

drugs.<br />

• There are several mechanisms to fix prices of orphan drugs.<br />

• The reimbursement procedure considers budget impact <strong>and</strong> sometimes<br />

cost-effectiveness. Often, dispensation <strong>for</strong> economic evaluation is given.<br />

<strong>Orphan</strong> drugs are fully or partially reimbursed by social insurance.<br />

• <strong>Orphan</strong> drugs are distributed through hospital <strong>and</strong> community pharmacies.<br />

• The orphan drug is prescribed by a specialist physician or a general<br />

practitioner. Conditions can be applied to the prescription of orphan drugs.<br />

• In the Netherl<strong>and</strong>s, hospitals may apply <strong>for</strong> full additional funding <strong>for</strong> orphan<br />

drugs that are prescribed within their institution through the policy<br />

measure. The additional temporally funding considers therapeutic value,<br />

cost prognosis <strong>and</strong> outcomes research – treatment of all patients need to be<br />

documented in a patient registry. After three years definitive listing<br />

considers: therapeutic value, budget impact, cost-effectiveness <strong>and</strong> efficient<br />

prescription. The appraisal will be based on these assessment criteria. There<br />

is no official threshold <strong>for</strong> the cost-effectiveness, a range is in place, the<br />

acceptable cost/QALY value will be balanced with other criteria depending<br />

on the individual case. In case of orphan drugs the therapeutic value, the<br />

severity of the disease <strong>and</strong> the efficient prescription will be important <strong>for</strong> the<br />

decision on definitive listing/ funding.<br />

4.7 SWEDEN nn<br />

4.7.1 Institutional context<br />

The institution in charge of providing in<strong>for</strong>mation on rare diseases is the Swedish<br />

National Centre <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong>.<br />

Reimbursement decisions are taken by the Dental <strong>and</strong> Pharmaceutical Benefits Board<br />

(DPBB), a governmental agency deciding whether or not a dental or pharmaceutical<br />

product will be subsidised by the State. 81<br />

There are specialised centres <strong>for</strong> rare diseases in each county (on a regional level)<br />

concentrating on clinical care, diagnosis <strong>and</strong> treatment. 60<br />

The Swedish National Board of Health <strong>and</strong> Welfare applies it own definition of a rare<br />

disease: “a disorder causing substantial disability <strong>and</strong> affecting fewer than 100 individuals per<br />

million population”. 9<br />

There is no policy <strong>for</strong> orphan drugs <strong>and</strong> there is no specific market access procedure<br />

<strong>for</strong> orphan drugs.<br />

4.7.2 Marketing Authorisation<br />

<strong>Orphan</strong> drugs obtain Marketing Authorisation through the centralised procedure at<br />

EMEA (see previous chapter).<br />

nn This chapter has been reviewed by Mr Karl Arnberg from the Dental <strong>and</strong> Pharmaceutical Benefits Board


60 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

4.7.3 Reimbursement<br />

The DPBB takes reimbursement decisions within a 180-day assessment period. The<br />

criteria are the same <strong>for</strong> orphan <strong>and</strong> non-orphan drugs being the cost-effectiveness of<br />

the product, the human value principle <strong>and</strong> the need <strong>and</strong> solidarity principle. The<br />

evidence asked is mostly the same clinical data as <strong>for</strong> the EMEA Marketing Authorisation<br />

procedure. Sometimes additional data is requested. 82<br />

The cost-effectiveness comprises a comparison of indirect <strong>and</strong> direct costs with the<br />

MAH’s health economics analysis. The human value principle implies equality of all<br />

persons <strong>and</strong> the need <strong>and</strong> solidarity principle implies that products that threat those<br />

with the greatest health needs take precedence. Even though cost-effectiveness is<br />

important <strong>for</strong> non-orphan drugs, the human value principle will prevail <strong>for</strong> both orphan<br />

<strong>and</strong> non-orphan drugs. 83 Cost-effectiveness must be proven, but as the threshold is<br />

higher <strong>for</strong> more severe diseases, a greater uncertainty is accepted if there is no possible<br />

way of acquiring data (e.g. due to a small patient group). 84<br />

The approved product is included on the List of Substitutable Products.<br />

Figure 4.13: Number of EMEA orphan drugs reimbursed in Sweden (2003-<br />

2008)<br />

Number of new <strong>Orphan</strong> <strong>Drugs</strong> per year<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

2003 2004 2005 2006 2007 2008<br />

Number of new <strong>Orphan</strong> <strong>Drugs</strong> per year Total number of <strong>Orphan</strong> <strong>Drugs</strong><br />

Source: Dental <strong>and</strong> Pharmaceutical Benefits Agency. Medicinal Products Database.<br />

. Accessed 28/4/2009.<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

Total number of <strong>Orphan</strong> <strong>Drugs</strong>


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 61<br />

4.7.4 Pricing<br />

The MAH is able to set the price freely, which will be approved by the DPBB following<br />

the abovementioned procedure. If accepted, the product will be included on the<br />

positive list of reimbursement. Medicinal products can also be sold without being<br />

included on the positive list, but patients will have to pay the full cost. This does not<br />

apply to products administered through hospitals: their price is the result of a<br />

negotiation between the MAH <strong>and</strong> the county councils. 83<br />

There is no price revision procedure: if a MAH wants to increase the price, the product<br />

will first have to be removed from the positive list <strong>and</strong> a new request will have to be<br />

introduced at the DPBB (substitution system). Or in order <strong>for</strong> the DPBB to approve a<br />

price increase <strong>for</strong> drugs not included in the substitution system, two conditions need to<br />

be fulfilled: 85<br />

1. The medicine in the application is an urgent therapeutic alternative as it is<br />

used to treat serious conditions which threaten the patient’s life <strong>and</strong> health.<br />

There are patients who risk being without similar treatment if the medicine<br />

disappears from the Swedish market.<br />

2. There is a considerable risk that the medicine will disappear from the<br />

Swedish market (or that the supply will decrease sharply), if the price<br />

increase is not approved.<br />

If the two conditions are fulfilled, the MAH will not have to withdraw from the positive<br />

list.<br />

4.7.5 Distribution<br />

4.7.6 Prescribing<br />

<strong>Orphan</strong> drugs are available through hospital <strong>and</strong> community pharmacies.<br />

The first prescription can be issued by the specialist physician or the general<br />

practitioner, but most patients are treated by a specialist. There are no conditions <strong>for</strong><br />

prescribing orphan drugs <strong>and</strong> there is no control mechanism. Reimbursement decisions<br />

do not differ between individuals. 84 Conditions on reimbursement can be imposed, but<br />

there are no general conditions specific <strong>for</strong> orphan drugs.<br />

The level of reimbursement is the same <strong>for</strong> all types of drugs. Up to 900 SEK (€ 81) oo of<br />

accumulated total cost of prescribed drugs the patient will bears the full cost. Between<br />

900 <strong>and</strong> 4,300 SEK the patient will pay a part of the costs <strong>and</strong> will receive the drugs free<br />

of charge once the accumulated total drugs cost has exceeded 4,300 SEK. An overview<br />

of the share of patient co-payment is given in the table 4.15. 83<br />

Figure 4.14: Patient co-payments in function of the accumulated total costs<br />

of prescribed drugs over 12 months in Sweden<br />

Accumulated total costs<br />

of prescribed drugs<br />

over 12 months<br />

Patient copayment<br />

Maximum<br />

accumulated<br />

patient outlay<br />

over 12 months<br />

≤ 900 SEK 100% 900 SEK<br />

901 – 1,700 SEK 50% 1,300 SEK<br />

1,701 – 3,300 SEK 25% 1,700 SEK<br />

3,301 – 4,300 SEK 10% 1,800 SEK<br />

≥ 4,300 SEK 0% 1,800 SEK<br />

Reimbursement is done by the Public Social Insurance.<br />

oo 1€ = 11,11SEK (20/4/2009)


62 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Key points<br />

• Sweden has in place specific centres of reference, but no policy measures<br />

<strong>and</strong> no research incentives on rare diseases/orphan drugs.<br />

• <strong>Orphan</strong> drugs are registered through the EMEA centralised procedure only.<br />

There is no legislation governing compassionate use or off-label use of<br />

orphan drugs.<br />

• There is free pricing of orphan drugs through a system of public<br />

procurement at the level of county councils.<br />

• The reimbursement procedure considers cost-effectiveness, but not budget<br />

impact because decisions are taken at the country level. <strong>Orphan</strong> drugs are<br />

fully reimbursed by social insurance.<br />

• <strong>Orphan</strong> drugs are available through hospital <strong>and</strong> community pharmacies.<br />

• The orphan drug is prescribed by a specialist physician or a general<br />

practitioner. No conditions are applied to the prescription of orphan drugs.<br />

4.8 UNITED KINGDOM pp<br />

4.8.1 Institutional context<br />

There is no specific funding <strong>for</strong> promoting the development of orphan drugs as these<br />

take place at a European level. Research projects to fund research networks <strong>for</strong> rare<br />

diseases were not identified.<br />

A distinction has to be made between the regulatory processes, pricing, HTA processes<br />

<strong>and</strong> the commissioning policies:<br />

• Regulatory processes: the medicine obtains a licence at EMEA level;<br />

• Pricing which is regulated by the Pharmaceutical Price Regulation Scheme<br />

(PPRS) 86 ;<br />

• HTA processes: Three HTA regional bodies provide guidance to the<br />

National Health Service on the use of health technologies based on<br />

appraisal of clinical <strong>and</strong> cost effectiveness evidence. 57 :<br />

o National Institute <strong>for</strong> Health <strong>and</strong> Clinical Excellence (NICE) <strong>for</strong><br />

Engl<strong>and</strong>. NICE produces guidance on public health, health technologies<br />

selected by the health ministers <strong>and</strong> clinical practice;<br />

o Scottish Medicines Consortium (SMC) <strong>for</strong> Scotl<strong>and</strong> which reviews all<br />

new medicines. SMC has developed a specific policy <strong>for</strong> orphan drugs;<br />

o All Wales Medicines Strategy Group (AWMSG) <strong>for</strong> Wales issues<br />

recommendations on drugs that have not been evaluated by NICE;<br />

• Commissioning: Commissioning in the National Health Service (NHS) is<br />

the process by which it is ensured that the health <strong>and</strong> care services<br />

provided most effectively meet the needs of the population qq .<br />

The cost effectiveness threshold used by NICE to make recommendations on the most<br />

appropriate use of medicines within the NHS is also applicable to orphan drugs.<br />

Following thresholds are applied: Below a most plausible ICER of £20,000/QALY,<br />

judgments about the acceptability of a technology as an effective use of NHS resources<br />

are based primarily on the cost-effectiveness estimate (point A in figure 4.15).<br />

pp This chapter has been reviewed by Ms Martina Garau of the Office of Health Economics; it describes<br />

mainly the situation in Engl<strong>and</strong>, unless mentioned differently in the text.<br />

qq The National Commisisoning Group (NCG) also has a top slice budget <strong>for</strong> therapies <strong>for</strong> very rare<br />

conditions. See also below.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 63<br />

Above a most plausible ICER of £20,000/QALY, judgments about the acceptability of<br />

the technology as an effective use of NHS resources are more likely to make more<br />

explicit reference to factors including:<br />

• the degree of uncertainty surrounding the calculation of ICERs<br />

• the innovative nature of the technology<br />

• the particular features of the condition <strong>and</strong> population receiving the<br />

technology<br />

• where appropriate, the wider societal costs <strong>and</strong> benefits.<br />

Above an ICER of £30,000/QALY, the case <strong>for</strong> supporting the technology on these<br />

factors has to be increasingly strong.(point B in figure 4.15) The reasoning <strong>for</strong> the<br />

Committee’s decision will be explained, with reference to the factors that have been<br />

87 88<br />

taken into account”.<br />

ICER st<strong>and</strong>s <strong>for</strong> incremental cost-effectiveness ratio being the extra cost that is paid <strong>for</strong><br />

each extra unit of health improvement gained by using the medicine, compared to the<br />

next most effective alternative. The ICER is measured in terms of the cost per QALY<br />

gained (quality-adjusted life year) by the intervention. A QALY gained is a year of life in<br />

good health a person might gain as a result of treatment.<br />

Medicinal products with an ICER higher than 30,000£ per QALY are usually not<br />

considered as cost effective. For a drug to be cost effective, it must deliver an additional<br />

QALY over <strong>and</strong> above treatments already available. Exceptions have been made <strong>for</strong><br />

orphan drugs: <strong>for</strong> example Imatinib <strong>for</strong> the treatment of myeloid leukaemia was<br />

approved at a cost of 48,000£ per QALY (being the highest cost ever accepted). 89 In<br />

January 2009, NICE published a supplementary advice indicating that life-extending<br />

medicines <strong>for</strong> end of life conditions affecting small populations can be recommended by<br />

NICE even when they exceed the cost-effectiveness threshold of £30,000/QALY. 87 This<br />

advice will influence decisions on treatments <strong>for</strong> rare cancers as they fall under end of<br />

life conditions, but not long-term chronic rare conditions. 57<br />

Figure 4.15: Incremental cost per QALY gained (ICER)<br />

Source: Rawlins MD, Culyer AJ. National Institute <strong>for</strong> Clinical Excellence <strong>and</strong> its value judgments.<br />

BMJ (Clinical research ed). 2004(329):224-7.<br />

A debate has been going on <strong>for</strong> some years on how to define orphan <strong>and</strong> ultra-orphan<br />

drugs. Recently, the Health Minister stated that there is no <strong>for</strong>mal classification of<br />

“ultra-orphan” drugs. The term has been used by NICE to indicate treatments <strong>for</strong><br />

conditions with a prevalence of less than one in 50,000 in the United Kingdom.


64 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

The National Commissioning Group (NCG) of the NHS selects diseases with less than<br />

400 cases. Its role is to commission services <strong>for</strong> the population of Engl<strong>and</strong> <strong>for</strong> a specific<br />

group of extremely rare conditions, which can include orphan drugs. 90<br />

4.8.2 Marketing Authorisation<br />

<strong>Orphan</strong> drugs obtain Marketing Authorisation through the centralised procedure at<br />

EMEA (see previous chapter).<br />

The body responsible <strong>for</strong> regulatory approval in the UK is the Medicines <strong>and</strong> Healthcare<br />

products Regulatory Agency (MHRA) which has not been involved with orphan drug<br />

Marketing Authorisation, to date.<br />

4.8.2.1 Compassionate use<br />

Prescription of unlicensed drugs is accepted if three conditions are fulfilled: 91<br />

• It is a bona fide unsolicited order;<br />

• The product is <strong>for</strong>mulated in accordance with the requirement of a<br />

doctor or dentist registered in the UK;<br />

• The product is <strong>for</strong> use by individual patients on their direct personal<br />

responsibility.<br />

There is not a specific procedure <strong>for</strong> compassionate use in the UK.<br />

4.8.2.2 Off-label use<br />

Off-label use is defined as the use of a licensed medicine <strong>for</strong> an unlicensed indication or<br />

administered via a different route. The medicine will be reimbursed if it is not<br />

particularly expensive. Otherwise, the reimbursement <strong>and</strong> the provision are monitored<br />

more strictly. The authority which monitors this is the Prescription Price Authority. 58<br />

4.8.3 Reimbursement<br />

4.8.3.1 Uptake of medicine on the market<br />

The UK has no system of reimbursement similar to the ones in other European<br />

countries as medicines are made available after launch <strong>and</strong> can in principle be prescribed<br />

by clinicians operating within the NHS as soon as the Marketing Authorisation is<br />

obtained. In practice, there are however mechanisms to control expenditure <strong>and</strong> a<br />

medicine can be appraised by one of the Health Technology Assessment (HTA) bodies<br />

(NICE, AWMSG, SMC), which issues guidance on its appropriate use within the NHS.<br />

The HTA body makes recommendations to the NHS about which drugs <strong>and</strong> treatments<br />

should be available.<br />

NICE evaluates orphan drugs using the same methods <strong>and</strong> decision criteria as <strong>for</strong> all<br />

technology appraisals, but a lower level of evidence may be accepted <strong>for</strong> orphan drugs. 92<br />

If NICE rejects a medicine, than the NHS clinicians cannot prescribe it. Technology<br />

appraisal consists of three steps: 93<br />

1. Scoping: what will be examined;<br />

2. Assessment of clinical <strong>and</strong> cost effectiveness: by means of a review of<br />

evidence <strong>and</strong> an economic evaluation (cost per QALY) conducted by an<br />

academic centre <strong>and</strong> the manufacturer/s.<br />

3. Appraisal of the assessment taking into account the opinion of consulters,<br />

commentators, clinical specialists <strong>and</strong> patient experts.<br />

While NICE assesses both old <strong>and</strong> new technologies, the SMC issues guidance on all<br />

newly licensed medicines, new indications <strong>and</strong> <strong>for</strong>mulations. 94 The AWMSG “appraises<br />

new high cost, cardiac <strong>and</strong> cancer medicines <strong>for</strong> which no NICE guidance is expected <strong>for</strong> at<br />

least twelve months”. 95<br />

The SMC has adopted a policy on orphan drugs according to which orphan drugs are<br />

appraised in the same way as normal drugs, but modifiers are considered. 96


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 65<br />

These modifiers are additional factors considered when orphan drugs are appraised,<br />

such as whether the drug:<br />

• treats a life threatening disease;<br />

• substantially increases life expectancy <strong>and</strong>/or quality of life;<br />

• can reverse, rather than stabilise, the condition;<br />

• or bridges a gap to a “definitive” therapy. 97<br />

The AWMSG’s criteria <strong>for</strong> ultra-orphan drugs are: 98<br />

• Degree of severity of the untreated disease, in terms of quality of life <strong>and</strong><br />

survival;<br />

• Whether the drug can reverse, rather than stabilise the condition;<br />

• Overall budget impact;<br />

• Whether the drug may bridge a gap to a “definitive” therapy which is<br />

currently in development;<br />

• The innovative nature of the drug.<br />

The assessment of the drugs takes into account the cost-effectiveness based on the<br />

price decided by the MAH 63<br />

As of April 2008, NICE had appraised only one EMEA-designated orphan drugs, which is<br />

imatinib <strong>for</strong> the treatment of gastro-intestinal stromal tumours <strong>and</strong> of chronic myeloid<br />

leukaemia. In both cases, the treatment was recommended <strong>for</strong> use. 99<br />

On the other h<strong>and</strong>, SMC had reviewed 28 orphan drugs. “Almost half of them were<br />

rejected (13), 12 were recommended <strong>and</strong> three were recommended <strong>for</strong> restricted use,<br />

i.e. <strong>for</strong> patient sub-group/s within the licensed indication”. 99<br />

The price of medicines funded by the NHS is included in a national list of tariffs, the<br />

British National Formulary (BNF). Funding takes place through the budget of the<br />

National Health Service.<br />

4.8.3.2 Commissioning<br />

Engl<strong>and</strong>, Wales <strong>and</strong> Scotl<strong>and</strong> have each developed specific funding mechanisms <strong>for</strong><br />

orphan drugs, which are broadly similar. In Engl<strong>and</strong>, health care services, including<br />

medicines, <strong>for</strong> very rare diseases are commissioned by the NCG. The NCG will assess<br />

diseases with an incidence of less than 400 cases. 44 On a regional level, services can be<br />

referred to the Specialised Commissioning Groups.<br />

The commissioning consists of an assessment of the health service needs <strong>and</strong> the<br />

current service provision <strong>for</strong> the local population. Based on this assessment, the<br />

Primary Care Trusts (PCTs) at local level identify what type <strong>and</strong> level of services need<br />

to be procured in the coming year from primary care services providers, such as<br />

General Practitioners or pharmacists, or from secondary care institutions, such as<br />

hospitals <strong>and</strong> mental health trusts. 55 The PCTs are responsible <strong>for</strong> the funding.<br />

4.8.4 Pricing<br />

The pricing mechanism is the same <strong>for</strong> orphan <strong>and</strong> non-orphan drugs.<br />

Prices are set freely by the MAHs, but have to meet the profit control criteria included<br />

in the PPRS. This scheme is an agreement between the Department of Health <strong>and</strong> the<br />

Association of the British Pharmaceutical Industry. 86<br />

There are two mechanisms <strong>for</strong> price revisions:<br />

• “flexible pricing where a price decrease or increase by the MAH is<br />

possible if there is new evidence or if a different indication is being<br />

developed (the flexible pricing mechanism will allow to have prices which<br />

better reflect the drug therapeutic value);<br />

• patient access schemes: early access to medicines which are not in first<br />

instance found to be cost <strong>and</strong> clinically effective by NICE”. 100


66 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

According to the 2009 PPRS agreement, MAHs are able to modulate the list price of<br />

their PPRS products by changes that equate to an overall level of 3.9% in 2009.<br />

Price revisions take place on an infrequent basis.<br />

4.8.5 Distribution<br />

4.8.6 Prescribing<br />

<strong>Orphan</strong> drugs are distributed through hospital pharmacies <strong>and</strong> specialist centres.<br />

The first prescription will be issued by the specialist physician. The prescription has to<br />

be consistent with the license.<br />

The prescription process is influenced, <strong>and</strong> there<strong>for</strong>e controlled, by the guidance, when<br />

available, of the HTA bodies on the use of medicines within the NHS.<br />

Differences in individual HTA decisions occur on a regional level as medicines are<br />

appraised by different HTA bodies (NICE, SMC <strong>and</strong> AWMSG) who can take different<br />

decisions.<br />

Key points<br />

• There are no policy measures <strong>and</strong> research incentives on rare<br />

diseases/orphan drugs in the UK. There are specific centres of reference <strong>for</strong><br />

some orphan drugs.<br />

• <strong>Orphan</strong> drugs are registered through the EMEA centralised procedure only.<br />

Specific legislation governs compassionate use <strong>and</strong> off-label use of orphan<br />

drugs.<br />

• Prices are set freely by the MAHs, but have to meet profit control criteria.<br />

• <strong>Orphan</strong> drugs are either fully or not reimbursed<br />

• The reimbursement procedure considers budget impact <strong>and</strong> costeffectiveness.<br />

<strong>Orphan</strong> drugs are fully reimbursed by the National Health<br />

Service.<br />

• <strong>Orphan</strong> drugs are available through hospital pharmacies.<br />

• <strong>Orphan</strong> drugs are prescribed by specialist physicians. The prescription<br />

process is influenced by the guidance of HTA bodies, if available.<br />

4.9 COMPARATIVE ANALYSIS<br />

Regulatory traits of the rare disease <strong>and</strong> orphan drug market in the six countries<br />

studied are presented in Figure 4.16.<br />

With respect to the institutional context, France, Italy, Sweden <strong>and</strong> (partly) the UK have<br />

dedicated centres of reference <strong>for</strong> orphan drugs <strong>and</strong> rare diseases. University medical<br />

centres fulfil this role in the Netherl<strong>and</strong>s. A similar situation applies <strong>for</strong> Belgium as<br />

mentioned above with four networks of centres that can be considered to partially fullfil<br />

the role of centres of reference. In addition to European measures to promote research<br />

<strong>and</strong> development of orphan drugs, France, Italy <strong>and</strong> the Netherl<strong>and</strong>s have implemented<br />

additional policy measures <strong>and</strong> research incentives <strong>for</strong> orphan drugs <strong>and</strong> rare diseases.<br />

<strong>Orphan</strong> drugs are registered through the EMEA centralised procedure in all six<br />

countries. Countries have introduced specific legislation governing compassionate use of<br />

orphan drugs, except <strong>for</strong> Sweden. Italy, the Netherl<strong>and</strong>s <strong>and</strong> the UK have implemented<br />

a procedure <strong>for</strong> off-label use of orphan drugs.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 67<br />

Prices of orphan drugs are subject to price fixing in all countries, except <strong>for</strong> Sweden (at<br />

the county level) <strong>and</strong> the UK. In France, Italy <strong>and</strong> the Netherl<strong>and</strong>s, prices are fixed with<br />

reference to, amongst other things, the price level in other EU countries. In the<br />

Netherl<strong>and</strong>s, maximum prices are fixed <strong>for</strong> orphan drugs. In order to maximize price<br />

competition, prices in Sweden are determined by a system of public procurement at the<br />

regional level. In the UK, orphan drug prices are set freely by the MAHs, but have to<br />

meet the profit control criteria.<br />

To gain reimbursement, <strong>for</strong>mal cost-effectiveness analysis is sometimes but not always<br />

per<strong>for</strong>med <strong>for</strong> orphan drugs in the countries studied. The budget impact of orphan<br />

drugs is considered in the reimbursement application in all countries, except <strong>for</strong><br />

Sweden. <strong>Orphan</strong> drugs are not always fully reimbursed in all countries studied. <strong>Orphan</strong><br />

drugs are fully reimbursed in Italy, Sweden, Belgium <strong>and</strong> the UK. France <strong>and</strong> the<br />

Netherl<strong>and</strong>s operate a mixed system of full or partial reimbursement.<br />

<strong>Orphan</strong> drugs are distributed through hospital pharmacies in all countries studied.<br />

Additionally, they are distributed through community pharmacies in Italy, France, the<br />

Netherl<strong>and</strong>s, <strong>and</strong> Sweden; they are also distributed through health authorities in Italy;<br />

<strong>and</strong> through specialist centres in the UK.<br />

<strong>Orphan</strong> drugs are initially prescribed by a specialist physician or a general practitioner in<br />

the Netherl<strong>and</strong>s <strong>and</strong> Sweden. The prescription is the exclusive responsibility of the<br />

specialist physician in Belgium, Italy <strong>and</strong> the UK. All countries studied impose conditions<br />

on prescribing orphan drugs, except <strong>for</strong> Sweden. In Italy, if an orphan drug is prescribed<br />

to a patient, the treatment must be registered in a national registry. Delivery of the<br />

drugs depends on provision of the data <strong>for</strong> the registration. This is also partly the case<br />

in Belgium <strong>for</strong> orphan drugs <strong>for</strong> which a CMDOD exists. In France, orphan drugs are<br />

reimbursed only if the rare disease is one of the indications. In the Netherl<strong>and</strong>s, health<br />

insurance funds have the right to impose additional prescribing conditions. In the UK,<br />

the prescription process of orphan drugs is influenced by the guidance of HTA bodies, if<br />

available.<br />

The number of available orphan drugs per country varies:<br />

Belgium 31 orphan drugs reimbursed, 2 not reimbursed but<br />

available (2008) rr<br />

France 35 orphan drugs (2007)<br />

Italy 23 molecules (2007)<br />

The Netherl<strong>and</strong>s 44 orphan drugs of which 36 are reimbursed (2008)<br />

Sweden 28 orphan drugs<br />

United Kingdom In<strong>for</strong>mation not available. All are in theory available,<br />

but not all are reimbursed.<br />

The highest availability of orphan drugs is achieved in the Netherl<strong>and</strong>s <strong>and</strong> France.<br />

rr See table 8.1 in annex <strong>for</strong> the full overview. <strong>Drugs</strong> available <strong>for</strong> compassionate use or available but not yet<br />

reimbursed are not included.


68 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Figure 4.16: Regulation governing rare disease <strong>and</strong> orphan drug markets<br />

Features<br />

INSTITUTIONAL CONTEXT<br />

Belgium France Italy Netherl<strong>and</strong>s Sweden UK<br />

Existence of centres <strong>for</strong> rare diseases/orphan drugs: No Yes Yes No Yes Yes<br />

Policy measures to promote development of orphan drugs: No Yes Yes Yes No No<br />

Incentives <strong>for</strong> research on rare diseases/orphan drugs: No Yes Yes Yes No No<br />

MARKETING AUTHORISATION<br />

Existence of national Marketing Authorisation procedure: No No No No No No<br />

Procedure <strong>for</strong> compassionate use of orphan drugs: Yes Yes Yes Yes No Yes<br />

Procedure <strong>for</strong> off-label use of orphan drugs: No No Yes Yes No Yes<br />

PRICING<br />

Pricing system:<br />

- Free market<br />

- Price fixing<br />

REIMBURSEMENT<br />

Third-party payer:<br />

Yes Yes Yes Yes<br />

Yes (county<br />

level)<br />

Yes (national<br />

level)<br />

- National Health Service Yes Yes<br />

- Social insurance Yes Yes Yes Yes<br />

Reimbursement based on cost-effectiveness: No Yes Yes Sometimes Yes Sometimes<br />

Reimbursement based on budget impact: Yes Yes Yes Yes No Yes<br />

Reimbursement level:<br />

- Full reimbursement Yes Yes Yes Yes Yes Yes<br />

- Partial reimbursement No Yes Yes<br />

DISTRIBUTION CHANNELS<br />

Delivery channels:<br />

- Hospital pharmacies Yes Yes Yes Yes Yes<br />

Yes


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 69<br />

Features Belgium France Italy Netherl<strong>and</strong>s Sweden UK<br />

- Community pharmacies Yes Yes Yes<br />

- Health authorities Yes<br />

- Internet<br />

- Other<br />

PRESCRIBING PROCESS<br />

Prescription by:<br />

- Specialist physician Yes Yes Yes Yes Yes<br />

- Nurse practitioner<br />

- General practitioner Yes Yes<br />

Existence of conditions <strong>for</strong> prescribing orphan drugs: Yes Yes Yes Yes No Yes


70 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

5 CRITICAL ASSESSMENT<br />

5.1 INTRODUCTION<br />

Chapter 4 has described the reimbursement procedure <strong>for</strong> orphan drugs in Belgium.<br />

Since 2002, the Drug Reimbursement Committee (DRC) of the NIHDI (National<br />

Institute <strong>for</strong> Health <strong>and</strong> Disability Insurance), the Belgian third-party payer, evaluates<br />

drug reimbursement requests based on multiple criteria: the therapeutic value, price<br />

<strong>and</strong> proposed reimbursement tariff, the importance of the drug in clinical practice, <strong>and</strong><br />

the budget impact of the drug. No economic evaluation of the orphan drug is required<br />

<strong>for</strong> reimbursement purposes.<br />

The aim of this chapter is to carry out a critical assessment of reimbursement request<br />

files of orphan drugs that have been submitted in Belgium since end 2001, the date that<br />

the new reimbursement procedure (not specific to orphan drugs) came into effect ss .<br />

First, a qualitative overview was conducted of the reimbursement dossiers of all orphan<br />

drugs focusing on the evidence submitted <strong>for</strong> each reimbursement criterion. Second, a<br />

number of orphan drugs were selected <strong>for</strong> an in-depth analysis. A critical assessment<br />

provided in the context of a drug reimbursement request <strong>for</strong> these orphan drugs was<br />

conducted <strong>and</strong> compared with the assessment report of the DRC (see point 5.4.2).<br />

Key points<br />

• This chapter conducts a qualitative overview of the reimbursement files of<br />

all Belgian orphan drugs focusing on the evidence submitted <strong>for</strong> each drug<br />

<strong>for</strong> each reimbursement criterion.<br />

• A critical assessment was carried out of the scientific evidence <strong>for</strong> 8 selected<br />

orphan drugs <strong>and</strong> compared with the assessment report of the DRC.<br />

5.2 METHODOLOGY<br />

5.2.1 Qualitative overview<br />

The qualitative overview was based on an examination of the reimbursement request<br />

files of orphan drugs submitted to the DRC. The following in<strong>for</strong>mation was extracted<br />

from the dossiers: description of the orphan drug; reimbursement status; therapeutic<br />

value <strong>and</strong> needs; budget impact; <strong>and</strong> number of registered indications. Each drug was<br />

identified in terms of its name, code according to the Anatomical Therapeutic Chemical<br />

(ATC) drug classification system 101 <strong>and</strong> supplier. The reimbursement status related to<br />

whether the dossier was an original application or a revision <strong>and</strong> whether<br />

reimbursement had been awarded. The quality of clinical evidence used to assess the<br />

therapeutic value of orphan drugs was evaluated by focusing on the number <strong>and</strong> design<br />

of clinical studies. The analysis also considered whether clinical studies had been<br />

published in peer-reviewed journals. The therapeutic needs <strong>for</strong> an orphan drug were<br />

analysed by taking into account its place in clinical practice (first- or second-line<br />

treatment) <strong>and</strong> whether any alternative treatment existed. The budget impact was<br />

determined by means of the number of patients <strong>and</strong> the cost per patient per year as<br />

reported in the reimbursement dossiers. Finally, the number of indications was<br />

reported <strong>for</strong> which the orphan drug was registered with the European Medicines<br />

Agency (EMEA) <strong>and</strong> <strong>for</strong> which the orphan drug sought reimbursement in Belgium.<br />

Companies need to submit a revised dossier to the DRC after 1.5 to 3 years following<br />

initial reimbursement approval (see chapter 4). Our analysis covered the finalised<br />

dossiers relating to the revised application of three orphan drugs, whose<br />

reimbursement was initially granted in 2004 <strong>and</strong> the initial application of 23 orphan<br />

drugs.<br />

ss KB 21/12/2001 tot vaststelling van de procedures, termijnen en voorwaarden inzake de tegemoetkoming<br />

van de verplichte verzekering voor geneeskundige verzorging en uitkeringen in de kosten van<br />

farmaceutische specialiteiten. Belgisch Staatsblad 29/12/2001.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 71<br />

5.2.2 In-depth analysis<br />

Eight orphan drugs were selected <strong>for</strong> an in-depth analysis. These cases were chosen to<br />

reflect the variety of reimbursement applications submitted to the DRC.<br />

5.2.2.1 Criteria <strong>for</strong> selection<br />

A panel of three experts agreed on the following selection criteria <strong>for</strong> cases (in<br />

descending order of importance):<br />

Criteria level 1:<br />

• Nature of disease: metabolism, oncology, toxicology, endocrinology,<br />

cardiovascular, hematology<br />

• Therapeutic value<br />

o Evidence published: yes/no<br />

o Number of studies <strong>and</strong> phase 1, 2, 3 or 4<br />

• Is it a first submission or a revision<br />

Criteria level 2:<br />

• Budget impact<br />

o Prevalence<br />

o Cost/patient/year<br />

Criteria level 3:<br />

• Supplier<br />

• Therapeutic need<br />

o 1 st or 2 nd line treatment<br />

o Alternative available: yes/no<br />

5.2.2.2 Choice of cases<br />

Applying the criteria <strong>and</strong> looking <strong>for</strong> the highest coverage <strong>for</strong> each criterion resulted in<br />

the following selection: Pedea®, Aldurazyme®; Fabrazyme®; Replagal®; Tracleer®;<br />

Trisenox®; Xagrid®; Zavesca®. Pedea® was a ‘negative’ case. The 7 other drugs turned<br />

out to be the ‘oldest’ cases.<br />

Together, these provide <strong>for</strong> a good spread over the different situations that can occur,<br />

over the natures of diseases possible, st<strong>and</strong> <strong>for</strong> a significant potential budget impact, <strong>and</strong><br />

are produced by various different manufacturers.<br />

Figure 5.1 Overview of the eight cases<br />

Aldurazyme® Fabrazyme® Replagal® Tracleer® Trisenox® Xagrid® Zavesca® Pedea®<br />

Nature of disease metabolic metabolic metabolic cardiovascular oncology hematology metabolic cardiovascular<br />

Evidence published no yes yes no yes yes yes No<br />

Phase study 1 double-blind 1 double-blind 2x2 1x3 1x2 1x1/2 1x1/2 6 RCTs<br />

RCT<br />

placebo<br />

2x3<br />

1x4<br />

First or revision 2 2 2 1 1 1 1 1<br />

Budget impact 12 /<br />

50-75 / 50-75 / 300 / 9 / 11000 / 90 / 200-300 /<br />

(prevalence /<br />

cost per patient<br />

per year)<br />

€40 000 €200 000 €200 000 €39 000 €25 000 €7 500 €94 000 €381<br />

supplier Genzyme Genzyme Shire EGT Actelion Cephalon Shire Actelion <strong>Orphan</strong><br />

Europe<br />

1st or 2nd line 1 1 1 1 2 2 2 1<br />

Alternative No Yes, Replagal Yes, Yes No Yes Yes Yes,<br />

available<br />

Fabrazyme<br />

Indomethacine


72 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Key points<br />

• The qualitative overview extracted in<strong>for</strong>mation about the orphan drug;<br />

reimbursement status; therapeutic value <strong>and</strong> needs; budget impact; <strong>and</strong><br />

number of registered indications from each reimbursement dossier.<br />

• A number of orphan drugs were selected <strong>for</strong> in-depth analysis. They were<br />

selected on the basis of the nature of disease, therapeutic value,<br />

reimbursement status, budget impact, supplier <strong>and</strong> therapeutic needs.<br />

5.3 QUALITATIVE OVERVIEW OF ALL REIMBURSEMENT<br />

DOSSIERS<br />

Between January 2002 <strong>and</strong> June 2008, reimbursement dossiers of 26 orphan drugs<br />

submitted to the DRC have been finalised. Reimbursement has been awarded to the<br />

majority of orphan drugs (22 out of 26 drugs). Table 9.2 in annex summarises<br />

reimbursement dossiers of all 26 drugs. Some of these have been re-submitted at a later<br />

date.<br />

The DRC’s advice was positive <strong>for</strong> 19 drugs. All these were approved by the Minister of<br />

Social Affairs.<br />

For one drug, there was no advice from the DRC, as no consensus could be reached.<br />

This drug was approved by the Minster of Social Affairs<br />

Two out of the six drugs <strong>for</strong> which the DRC’s advice was negative, were granted<br />

reimbursement by the Minister.<br />

For two of these three drugs it appears from the dossiers that both the DRC <strong>and</strong> the<br />

pharmaceutical company proposed a number of elements <strong>for</strong> negotiation - including a<br />

price decrease, employment opportunities, restrictions on the size of the patient<br />

population, the funding of diagnostic tests by the company, a reduction of the dosage -<br />

which may have played a role in awarding reimbursement.<br />

In the case of the third orphan drug, EMEA had granted an initial conditional marketing<br />

authorisation subject to the condition that the pharmaceutical company carried out<br />

additional clinical studies <strong>and</strong> submitted a new registration application to EMEA. In<br />

Belgium, reimbursement was granted to this drug, even though the DRC noted that<br />

there was insufficient evidence of the effectiveness of the drug in daily clinical practice<br />

<strong>and</strong> in the long-term. Following a new registration application, the DRC will revisit the<br />

reimbursement application. The provisional award of reimbursement in return <strong>for</strong> an<br />

engagement to undertake further clinical research amounts to a public subsidy <strong>for</strong><br />

clinical research. It could be argued that this creates an uneven playing field <strong>for</strong> clinical<br />

research between pharmaceutical companies.<br />

The rationale <strong>for</strong> not granting reimbursement to four orphan drugs may be related to<br />

the high cost of the orphan drug in comparison with alternative drugs or the existence<br />

of other non-orphan indications of the drug.<br />

Using the first level of the ATC drug classification system, orphan drugs mainly related<br />

to ‘L Antineoplastic <strong>and</strong> immunomodulating agents’ (10 drugs) <strong>and</strong> ‘A Alimentary tract<br />

<strong>and</strong> metabolism’ (9 drugs); but also included ‘C Cardiovascular system’ (2 drugs); ‘V<br />

Various’ (2 drugs); ‘G Genitor-urinary system <strong>and</strong> sex hormones’ (1 drug); ‘H Systemic<br />

hormonal preparations, excluding sex hormones <strong>and</strong> insulins’ (1 drug); <strong>and</strong> ‘N Nervous<br />

system’ (1 drug).<br />

The evidence of therapeutic value included in the reimbursement dossier was similar to<br />

the evidence submitted to EMEA <strong>for</strong> registration purposes. This may reflect the short<br />

time period (an average of 130 days according to NIHDI/INAMI data) between EMEA<br />

registration approval <strong>and</strong> submission of the reimbursement application to the DRC in<br />

Belgium. The reimbursement dossier of one orphan drug included an additional clinical<br />

study that had not been available at the time of registration with EMEA.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 73<br />

In general, the evidence of therapeutic value was limited, with evidence derived from<br />

few clinical studies.<br />

The methodological design of studies varied considerably, with clinical evidence derived<br />

from double-blind r<strong>and</strong>omised controlled trials, open-label studies <strong>and</strong> case series. Ten<br />

dossiers included clinical evidence from double-blind r<strong>and</strong>omised controlled trials <strong>for</strong><br />

orphan drugs relating to various ATC drug classes including ‘A Alimentary tract <strong>and</strong><br />

metabolism’, ‘C Cardiovascular system’, ‘G Genitor-urinary system <strong>and</strong> sex hormones’,<br />

‘L Antineoplastic <strong>and</strong> immunomodulating agents’ <strong>and</strong> ‘G Genitor-urinary system <strong>and</strong> sex<br />

hormones’. Clinical studies of 13 orphan drugs had been published in peer-reviewed<br />

journals.<br />

<strong>Orphan</strong> drugs were positioned as first-line treatment (11 drugs) or second-line<br />

treatment (10 drugs) or both (4 drugs). There appears to be a therapeutic need <strong>for</strong><br />

some orphan drugs in the absence of alternative treatments. However, alternative<br />

treatments were available <strong>for</strong> 15 orphan drugs. In this respect, it should be noted that<br />

some orphan drugs shared common indications, i.e. Nexavar ® <strong>and</strong> Sutent ® <strong>for</strong> advanced<br />

renal cell carcinoma; Fabrazyme ® <strong>and</strong> Replagal ® <strong>for</strong> Fabry disease; Revatio ® , Tracleer ®<br />

<strong>and</strong> Thelin ® <strong>for</strong> pulmonary arterial hypertension; Glivec ® , Sprycel ® <strong>and</strong> Tasigna ® <strong>for</strong><br />

chronic myeloid leukaemia; Ceplene ® , Revlimid ® <strong>and</strong> Thalidomide ® <strong>for</strong> multiple<br />

myelome.<br />

In the absence of Belgian methodological guidelines to conduct a budget impact analysis,<br />

analyses included in reimbursement dossiers were generally simplistic. The number of<br />

patients <strong>and</strong> drug market share in Belgium were estimated or assumed by the<br />

pharmaceutical company. Budget impact analyses considered drug reimbursement tariffs<br />

rather than public prices. No dossier took into account the fact that the potential<br />

reimbursement of the orphan drug is likely to influence the market share of <strong>and</strong> the<br />

number of patients using alternative drugs or treatments. Potential savings are nearly<br />

never mentioned. Analyses were limited to examining the impact of drug costs <strong>and</strong> did<br />

not consider total treatment costs. One can assume this is not done as the cost of the<br />

drug is dominant in the treatment cost, <strong>and</strong> other costs like consultations or tests, are<br />

marginal in comparison.<br />

In general, reimbursement was sought in Belgium <strong>for</strong> the indication registered with<br />

EMEA. For three drugs, a reimbursement application was submitted <strong>for</strong> one of two<br />

indications registered with EMEA. If a pharmaceutical company submits multiple<br />

reimbursement dossiers relating to different indications of the orphan drug rather than<br />

one dossier relating to all indications, the DRC assesses the budgetary impact <strong>for</strong> an<br />

individual indication, but is not able to assess the total budgetary impact spanning all<br />

indications of the orphan drug.<br />

Key points<br />

• Reimbursement is awarded to the majority of orphan drugs.<br />

• In addition to the official criteria used by the DRC, other arguments such as<br />

price, employment, patient population, funding of diagnostic tests by the<br />

company may play a role in the reimbursement decision of the Minister.<br />

• The provisional award of reimbursement to one orphan drug in return <strong>for</strong><br />

an engagement to undertake further clinical research in effect amounts to a<br />

public subsidy <strong>for</strong> clinical research.<br />

• Decisions of not granting reimbursement to some orphan drugs were<br />

related to the high cost of the orphan drug in comparison with alternative<br />

drugs or the existence of other non-orphan indications of the drug.<br />

• It is possible to derive evidence of therapeutic value from double-blinded<br />

r<strong>and</strong>omized controlled trials.<br />

• There appears to be a therapeutic need <strong>for</strong> some orphan drugs in the<br />

absence of alternative treatments.<br />

• Budget impact analyses were simplistic <strong>and</strong> there is a need <strong>for</strong> principles of<br />

good practice <strong>for</strong> budget impact analyses.


74 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

• The DRC needs to consider the total budget impact of successive<br />

reimbursement dossiers of an orphan drug relating to different indications.<br />

5.4 IN-DEPTH ANALYSIS OF 15 SELECTED REIMBURSEMENT<br />

DOSSIERS<br />

5.4.1 Comparison of the evaluations by EMEA<br />

5.4.1.1 Objective<br />

One of the objectives of the <strong>KCE</strong> project was to document the different steps leading<br />

to approval <strong>and</strong> reimbursement of orphan drugs in Belgium. The data on which<br />

decisions are based, are provided to EMEA by the company in the <strong>for</strong>m of a Marketing<br />

Authorisation application file (Common Technical Dossier, CTD). If regulatory approval<br />

is obtained a European Public Assessment Report (EPAR) <strong>and</strong> the Summary of Product<br />

Characteristics (SPC) are made public by EMEA. In order to obtain reimbursement <strong>for</strong><br />

the approved drug the company provides data to the Belgian agency deciding on<br />

reimbursement (NIHDI).<br />

In order to get a feeling <strong>for</strong> the possible redundancy of the local evaluation it was<br />

checked whether the same study data sets were provided to EMEA <strong>and</strong> NIHDI, <strong>and</strong><br />

whether the EPAR could be considered an alternative <strong>for</strong> the local evaluation (<strong>and</strong> if not<br />

how, it could be improved to also serve this purpose).<br />

5.4.1.2 Methodology<br />

First, a list of EMEA approved orphan drug indications was compiled based on either<br />

cases selected <strong>for</strong> the <strong>KCE</strong> study or recent approvals. The approval could be under<br />

exceptional circumstances or not. The list is given in annex 8.6.<br />

For each drug we focused on the first EMEA approval of orphan indication(s). We<br />

focused on clinical efficacy <strong>and</strong> only on the primary endpoint. This is a limitation of the<br />

study. Evaluating benefits <strong>and</strong> risks of an orphan drug in a specific indication involves<br />

much more than just looking at a primary endpoint. However the primary endpoint has<br />

the advantage that the method of analysis is (or should be) pre-defined in the study<br />

protocol <strong>and</strong> the statistical analysis plan of the sponsor.<br />

In case Marketing Authorisation had been granted under exceptional circumstances<br />

without study <strong>and</strong> demonstration of benefit based on clinical endpoints, the CHMP<br />

requested phase 4 clinical trial was considered instead, which was per<strong>for</strong>med in order<br />

to obtain a normal Marketing Authorisation.<br />

The pivotal clinical efficacy trials were identified <strong>and</strong> their pre-defined primary endpoint,<br />

as well as the result obtained. These steps were followed separately <strong>for</strong> the three data<br />

sources <strong>and</strong> compared.<br />

1. The Marketing Authorisation application file (common technical document<br />

(CTD part 2 (2.7.3), clinical summary which include the clinical summary or<br />

expert report, tabular <strong>for</strong>mats, study synopsis (the full ICH study reports<br />

were not checked).<br />

2. The European Public Assessment Report (EPAR) <strong>and</strong> the SPC.<br />

3. The file submitted by the company to the Belgian NIDHI <strong>for</strong> obtaining<br />

reimbursement.<br />

Data sources 1 <strong>and</strong> 2 were compared first. The CTD part 2.7.3 of the EMEA Marketing<br />

Authorisation application file was made available <strong>for</strong> review during a visit at EMEA in<br />

FEB 2009, under confidentiality. The EPAR reflecting the first orphan indication approval<br />

was in most cases available from the EMEA website or was made available <strong>for</strong> review<br />

during a visit at EMEA in FEB 2009, under confidentiality.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 75<br />

5.4.1.3 Results <strong>for</strong> comparison of CTD part 2.7.3 <strong>and</strong> EPAR<br />

In all of the 15 drug-indication pairs reviewed we identified the same pivotal trials <strong>and</strong> in<br />

most cases no differences were seen <strong>for</strong> the primary endpoint results between the<br />

CTD part 2.7.3 <strong>and</strong> the EPAR/SPC.<br />

As these are orphan drugs, <strong>for</strong> the vast majority of the 15 cases there is only a single<br />

pivotal trial with clinical endpoints.<br />

In two out of the 15 cases the results of the main statistical test as pre-defined in the<br />

study protocol <strong>for</strong> analysis of the primary endpoint was not mentioned in the<br />

EPAR/SPC, but instead only the result of an alternative statistical method was taken<br />

<strong>for</strong>ward by the assessors <strong>and</strong> communicated as a measure of risk reduction. In both<br />

cases the effect of treatment using the pre-defined main statistical method was not<br />

significant. In both cases the alternative statistical method provided a p-value that was<br />

smaller than the prospective analytic method, <strong>and</strong> was numerically less than 0.05 in one<br />

case. The meaning of such alternative analyses is uncertain, given both the post-hoc<br />

nature <strong>and</strong> the multiplicity of analyses. It must be stated that these observations refer to<br />

a period in time when no templates were in use <strong>for</strong> the CHMP assessment report.<br />

Structure <strong>and</strong> level of detail was left to the discretion of the rapporteurs. In 2002, <strong>and</strong><br />

following a major revision in 2004, the CHMP has adopted new templates <strong>for</strong> the<br />

assessment reports, including detailed guidance <strong>and</strong> structure in line with internationally<br />

agreed st<strong>and</strong>ards of scientific publications based on the CONSORT statement (The<br />

Lancet 2001; 357: 1191-94). Such templates are currently in use throughout the<br />

scientific assessment <strong>and</strong> have improved the assessment reports.<br />

In case of ongoing trials, the differences between the CTD <strong>and</strong> the EPAR in results <strong>for</strong><br />

the primary efficacy variables was explained by additional in<strong>for</strong>mation (more patients,<br />

longer follow-up) which became available at EMEA during that part of the review<br />

process which ends with drug approval <strong>and</strong> publication of the EPAR. This additional<br />

in<strong>for</strong>mation was on file at EMEA but was not available <strong>for</strong> verification during the EMEA<br />

visit. An additional visit <strong>for</strong> checking these items was not considered necessary by <strong>KCE</strong>.<br />

In case results of new studies are provided by the applicant after publication of the first<br />

EPAR it is not always possible to find the results <strong>for</strong> the primary endpoint at the EMEA<br />

website. This observation is in agreement with EMEA policies: only certain types of<br />

variations trigger a revision of the EPAR, such as variations of the therapeutic indication.<br />

5.4.2 Comparison of the studies mentioned in the NIHDI file, the EMEA file<br />

<strong>and</strong> EPAR<br />

A comparison has been made between the primary endpoints of studies mentioned in<br />

the NIHDI file (being the company’s application sent to NIHDI), the company’s<br />

application sent to EMEA <strong>and</strong> the in<strong>for</strong>mation contained in the EPAR. This comparison<br />

was per<strong>for</strong>med <strong>for</strong> fourteen orphan drugs <strong>and</strong> fifteen indications.<br />

• In six cases the in<strong>for</strong>mation provided in all three documents was the<br />

same.<br />

• One NIHDI file only contained the main study, not the supportive nor the<br />

extension study.<br />

• In two cases, the NIHDI file contains fewer studies than the EMEA file <strong>and</strong><br />

EPAR. For the first drug, of the two studies contained in the EMEA file<br />

<strong>and</strong> EPAR, the study phase I/II is not mentioned in the NIHDI file. As <strong>for</strong><br />

the other study, no numbers are given but the explanation is in line with<br />

the one of the EPAR. For the second drug, one phase II study with a<br />

primary endpoint of p=0.42 is not mentioned.<br />

• In one case the two studies of the NIHDI file correspond to the studies in<br />

the EMEA file <strong>and</strong> the EPAR. Nevertheless, the results of the second<br />

study are taken from the SPC (Summary of Product Characteristics)<br />

produced by EMEA. A negative study listed in the EMEA file is not<br />

mentioned in the NIHDI file.


76 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

• In one case the NIHDI file does not give figures, but explains the results,<br />

corresponding to the figures in the EMEA file <strong>and</strong> EPAR.<br />

• Two NIHDI files contain one additional study compared to the EMEA file<br />

<strong>and</strong> EPAR. This can be explained by the fact that the study started after or<br />

that the study’s first results were published after the EMEA procedure<br />

ended.<br />

• For one orphan drug, the studies are the same in all documents, but the<br />

results differ. This can be due to the fact that the study covered a long<br />

time-period <strong>and</strong> that results were measured at several times.<br />

• In one case the NIHDI file concludes the study showed a positive effect,<br />

while the EPAR states that “the data are too limited to conclude on the<br />

appropriate dosing in children”.<br />

In general, it can be said that the sponsor’s application files sent to NIHDI contain<br />

similar but not always the same data as the ones provided to EMEA <strong>and</strong> to be found in<br />

the EPAR. Differences are, when they occur, relatively small, although it is observed that<br />

if they occur, they are always to the advantage of the product. It is uncertain whether<br />

this has had an impact on the reimbursement decisions.<br />

Key points<br />

• In most cases, no differences were seen between the CTD part 2.7.3. <strong>and</strong> the<br />

EPAR/SPC in terms of primary endpoint results.<br />

• In two cases, an alternative statistical method was reported in the<br />

EPAR/SPC than the statistical test pre-defined in the study protocol.<br />

• In case of ongoing trials, differences between CTD <strong>and</strong> EPAR could be<br />

explained by additional in<strong>for</strong>mation becoming available during the review<br />

process.<br />

• Generally, the companies’ drug reimbursement request files sent to NIHDI<br />

contained similar but not always identical in<strong>for</strong>mation as the ones provided<br />

to EMEA <strong>and</strong> to be found in the EPAR. Small differences observed were<br />

always to the advantage of the product.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 77<br />

6 BUDGET IMPACT ANALYSIS<br />

6.1 METHODOLOGY<br />

The budget impact analysis of the orphan drugs on the Belgian health care budget is<br />

per<strong>for</strong>med based on the situation at the end of 2008.<br />

The analysis is split into two parts:<br />

• an estimate of the budget impact at the end of 2008 in Belgium;<br />

• the development <strong>and</strong> application of scenarios <strong>for</strong> the future to estimate<br />

budget impacts in the short <strong>and</strong> medium term.<br />

The estimate at the end of 2008 has been done combining all potential in<strong>for</strong>mation<br />

sources which is described in section 6.2 below. Most of these sources provide partial<br />

in<strong>for</strong>mation which leaves a high level of uncertainty.<br />

Forecasts are based on scenarios that are described in section 6.3. The starting point<br />

<strong>for</strong> these <strong>for</strong>ecasts is the estimate made <strong>for</strong> 2008.<br />

6.2 BUDGET IMPACT IN BELGIUM AT THE END OF 2008<br />

At the end of 2008, 31 different orphan drugs were approved <strong>for</strong> reimbursement in<br />

Belgium (of which 30 since 2003 when the Belgian orphan drug legislation was<br />

implemented). These 31 drugs correspond to 35 different indications.<br />

Figure 6.1 : Number of orphan drugs reimbursed per year in Belgium<br />

Approved in:<br />

1999 1<br />

2003 1<br />

2004 6<br />

2005 3<br />

2006 2<br />

2007 7<br />

2008 11<br />

Total orphan drugs 31<br />

More than half of the 31 drugs were approved in the last 24 months.


78 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Figure 6.2 : Number of orphan drugs reimbursed per year <strong>and</strong> total over the<br />

years 1999-2008 in Belgium<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

1999 2003 2004 2005 2006 2007 2008<br />

Number of <strong>Orphan</strong> <strong>Drugs</strong> per year Total <strong>Orphan</strong> <strong>Drugs</strong><br />

Figure 6.3 provides an estimate of the budget impact <strong>for</strong> all drugs approved by the end<br />

of 2008.<br />

These estimates are tentative, given the high degree of uncertainty of some variables.<br />

The reality lies probably in a bracket from -30 % of the calculated estimate to +30 %.<br />

For 2008, the real budget impact can there<strong>for</strong>e be located between 50 <strong>and</strong> 85 million<br />

Euro.<br />

The first source of in<strong>for</strong>mation is the Ministerial Decree which always includes an<br />

estimate of budget impact. This published in<strong>for</strong>mation 102 is based on the original file<br />

submitted by the sponsor to obtain reimbursement. The estimate made by the industry<br />

is always reviewed during the approval process, <strong>and</strong> sometimes a new (different)<br />

estimate is made by the DRC.<br />

The estimates provided in the Ministerial Decree are based on assumptions regarding<br />

some variables <strong>and</strong> are thus uncertain:<br />

• The number of patients is unknown, <strong>and</strong> simply applying the prevalence<br />

figures systematically leads to overestimates because not all patients will<br />

be treated.<br />

• In most cases, some time is needed to actually identify patients who may<br />

use the drug: the uptake of the medicine takes time (a few years).<br />

• Not all patients actually consume the doses as defined by the industry.<br />

There can be various reasons <strong>for</strong> this, but in practice, this will lead to a<br />

lower budget impact than <strong>for</strong>ecasted.<br />

35<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 79<br />

Various other sources were used to make the estimate <strong>and</strong> cross-check:<br />

• Figures published by the NIHDI based on their internal in<strong>for</strong>mation. These<br />

figures were made public at a hearing at the Federal Parliament in<br />

February 2009 <strong>and</strong> are also part of the MORSE report. 103 These include<br />

the number of patients that filed a dem<strong>and</strong> to “colleges” <strong>for</strong> approval of<br />

reimbursement.<br />

• Figures available at the FPS Economy. The FPS Economy is in charge of<br />

approving pricing, <strong>and</strong> collects on a yearly basis the turnover in<strong>for</strong>mation<br />

directly from the industry. The in<strong>for</strong>mation obtained covered 2007 <strong>and</strong><br />

was <strong>for</strong> a relatively small number of drugs.<br />

• Revision files: various drugs had to submit files <strong>for</strong> revisions, whether as a<br />

planned revision <strong>for</strong> drugs having been more than three years on the<br />

market, or because they asked <strong>for</strong> an extension (e.g. a new dose). These<br />

files <strong>and</strong> the published Ministerial Decrees, include more recent<br />

in<strong>for</strong>mation on the budget impact than was available in the original files.<br />

• In<strong>for</strong>mation from the SSF: the SSF is used to obtain reimbursement <strong>for</strong><br />

individual patients between the Marketing Authorisation <strong>and</strong> the<br />

reimbursement decision by the CTG.<br />

• IMS figures.<br />

All estimates are based on a calculation based on the number of patients (linked to the<br />

prevalence) <strong>and</strong> the average cost of a treatment of a patient. The table below there<strong>for</strong>e<br />

includes the in<strong>for</strong>mation on prevalence, number of patients <strong>and</strong> average cost. A scoring<br />

was also included as to the reliability of the estimate:<br />

• Score A = both the number of patients <strong>and</strong> the average cost can be<br />

considered as fairly reliable estimates;<br />

• Score B = either the number of patients or the average cost can be<br />

considered as highly uncertain;<br />

• Score C = both the number of patients <strong>and</strong> the average cost can be<br />

considered as uncertain.<br />

This estimate should be considered as an operational exercise rather than scientific as it<br />

combines different types of in<strong>for</strong>mation sources: <strong>for</strong>ecasts <strong>for</strong> the drugs recently<br />

launched <strong>and</strong> real figures <strong>for</strong> drugs that are longer on the market.


<strong>Orphan</strong> drug<br />

80 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Figure 6.3 : Overview of the estimated budget impact of orphan drugs in<br />

Belgium<br />

Reimbursed since<br />

Nature of disease<br />

(O= oncological, NO<br />

= not oncological)<br />

Number of Patients<br />

treated<br />

Cost/patient / year in<br />

000 €<br />

Total estimated cost<br />

to budget 2008 in 000<br />

€<br />

Aldurazyme® 1/8/04 NO 9 312 3,600 A<br />

Atriance® 1/6/08 O 24 23 / adult; 14<br />

/ child<br />

Reliability of estimate<br />

160 B<br />

Busilvex® 1/10/08 NO ~44 4.6 205 B<br />

Carbaglu® 1/9/06 NO 14 -1 085 1,106 A<br />

Duodopa® 1/3/07 NO 73 49 4,000 A<br />

Elaprase® 1/1/08 NO 8 300 1,600 B<br />

Evoltra® 1/7/08 O ~10 64 200 C<br />

Exjade® 1/8/07 NO ~1 080 Varies 3,000 B<br />

Fabrazyme® 1/8/04 NO 48 195 7,500 A<br />

Glivec® As drug cat 2<br />

(1/7/2003)<br />

O 120 31- 48 4,800 A<br />

Increlex® 1/8/08 NO 1 40 40 A<br />

Lysodren® 1/1/08 O 36 6.5 167 C<br />

Myozyme® 1/5/07 NO 23-33 176 / child; 411 /<br />

<strong>for</strong> adult<br />

7,800 B<br />

Naglazyme® 1/12/08 NO 0 376 0 A<br />

Nexavar® 1/4/07 O 215 24 weeks: 15<br />

30 weeks: 19<br />

52 weeks: 33<br />

3,700 B<br />

Orfadin® 1/7/06 NO 14 50 -100 1,200 B<br />

Replagal® 1/8/04 NO Cost already counted in Fabrazyme B<br />

Revatio® 1/6/07 NO 70-142 7 - 26 1,500 B<br />

Revlimid® 1/4/08 O 60 5,500 B<br />

Savene® 1/9/07 NO 29 10 150 C<br />

Somavert® 1/4/04 NO 70 per year 1,600 C<br />

Sprycel® 1/9/07 O 85-900 4,800 B<br />

Sutent® 1/4/07 O GIST: 73<br />

mRCC: 180-<br />

240<br />

GIST: 6<br />

mRCC: 21<br />

3,000 C<br />

Tasigna® 1/9/08 O Cost counted with Sprycel B<br />

Thelin® 1/1/08 NO 50 32 3,900 A<br />

Torisel® 1/12/08 O 0 30 0 A<br />

Tracleer® 1/8/04 NO 358 37 4,700 B<br />

Trisenox® 1/11/05 O 4 37 275 A<br />

Xagrid® 1/11/05 NO 320 7 1,500 A<br />

Zavesca® 1/9/05 NO 2 93 200 A<br />

TOTAL 66,203


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 81<br />

Comments regarding this estimated budget impact:<br />

• One orphan drug has not been included because of absence of<br />

in<strong>for</strong>mation (Thalidomide®).<br />

• The estimated cost is only the cost of the drug, not of the total<br />

treatment.<br />

• Savings are not taken into account. For orphan drugs in the category “no<br />

alternative”, the potential saving is linked to treatment of symptoms of the<br />

disease. For orphan drugs in the category “significant benefit”, the saving<br />

is the alternative treatment.<br />

• The costs of the orphan drugs reimbursed through the SSF are not<br />

included in the table above, as they are not yet known <strong>for</strong> 2008 <strong>and</strong> cover<br />

orphan drugs that are not part of the “official list” of reimbursed drugs.<br />

(The SSF reimbursing only drugs not (yet) reimbursed under the normal<br />

scheme). For 2007, this cost was near to € 4 million, but one drug<br />

(Myozyme®) accounted <strong>for</strong> € 3.5 million.<br />

The total estimated cost to the NIHDI budget of € 66.2 million corresponds to more<br />

than 5 % of total hospital drugs budgets in 2008.<br />

6.3 BUDGET IMPACT FORECAST<br />

Three scenarios are applied to estimate the future budget impact: a conservative<br />

scenario (low growth/cost), a realistic scenario (best estimate) <strong>and</strong> a higher growth/cost<br />

scenario.<br />

These scenarios are based on following variables:<br />

• An estimate of the average number per year of orphan drugs that will<br />

obtain a Marketing Authorisation at the European level.<br />

• An estimate of the number of drugs per year that will obtain a positive<br />

reimbursement decision in Belgium.<br />

• The average cost per patient per year per drug.<br />

The applied estimate of the average number of orphan drugs that obtain a Marketing<br />

Authorisation is based on the past. At the end of 2008, 48 orphan drugs had obtained a<br />

Marketing Authorisation (Figure 6.4.).<br />

Forecasting the number of Marketing Authorisations can be based on the evolution of<br />

the number of <strong>Orphan</strong> Drug Designations granted by the EC.


82 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Number of approved MA per year<br />

Figure 6.4 : Number of approved Marketing Authorisations per year <strong>and</strong><br />

total number of Marketing Authorisations over the years 2001-2008<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

2001 2002 2003 2004 2005 2006 2007 2008<br />

Number of approved MA per year Total number of MA per year<br />

[Source]: DG Enterprise EC. Register of designated <strong>Orphan</strong> Medicinal Products.<br />

. 11/3/2009.<br />

The figure below gives an overview of this evolution over the years since the legislation<br />

on orphan drugs exists.<br />

Figure 6.5 : Overview of <strong>Orphan</strong> Designations 2000-2008<br />

Year Applications<br />

submitted<br />

Positive<br />

COMP<br />

Opinions<br />

Applications<br />

withdrawn<br />

Final negative<br />

COMP Opinions<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

0<br />

Total number of MA per year<br />

Designations<br />

granted by the<br />

Commission<br />

2008 119 86 31 1 73<br />

2007 125 97 19 1 98<br />

2006 104 81 20 2 80<br />

2005 118 88 30 0 88<br />

2004 108 75 22 4 72<br />

2003 87 54 41 1 55<br />

2002 80 43 30 3 49<br />

2001 83 64 27 1 64<br />

2000 72 26 6 0 14<br />

Total 896 614 226 13 593<br />

[Source]: Committee <strong>for</strong> <strong>Orphan</strong> Medicinal Products. January 2009 Plenary Meeting, Monthly<br />

Report. EMEA. Doc. Ref.: EMEA/COMP/694107/2008. 7 January 2009. Available from<br />

[Last accessed: 10/3/2009].


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 83<br />

A total of 593 designations were granted by the end of 2008.<br />

Both the designations <strong>and</strong> the Marketing Authorisations seem to have reached a “cruise<br />

speed”, also in comparison with the situation in the USA. The estimate is there<strong>for</strong>e that<br />

there will be an average increase of at least 10 drugs per year. This figure also<br />

corresponds to expert opinions <strong>and</strong> the expectation from EMEA. There are no signs at<br />

this stage that drugs might be taken off the market. This can however be expected to<br />

happen in the longer term, <strong>for</strong> example when new therapies are introduced that replace<br />

existing orphan drugs. This has not been taken into account in the <strong>for</strong>ecast as it<br />

probably will not have a significant effect in the next five years.<br />

• Realistic scenario: net increase of 10 new orphan drugs / year<br />

• Low growth scenario: net increase of 8 orphan drugs per year<br />

• High growth scenario: net increase of 12 orphan drugs per year<br />

Most of the orphan drugs that obtain a Marketing Authorisation are getting (after a<br />

delay) a positive reimbursement decision in Belgium. This has been the experience up to<br />

now, <strong>and</strong> there<strong>for</strong>e a transfer ratio of 90 % (9 out of 10 orphan drugs) has obtained a<br />

positive reimbursement decision in Belgium. This is valid <strong>for</strong> all the scenarios.<br />

• Realistic scenario: transfer ratio of 90 %<br />

• Low growth scenario: transfer ratio of 80 %<br />

• High growth scenario: transfer ratio of 100 %<br />

The average cost of a reimbursed drug over 2008 is estimated at € 2.135 million Euro.<br />

This is much higher than in 2007 when it was 1.6 million. The average of € 2.135 million<br />

is probably too low, as more than one in three orphan drugs were approved during<br />

2008, <strong>and</strong> were introduced in the course of the year. Their budget impact will be higher<br />

in 2009. The € 2.135 million is on the other h<strong>and</strong> a high average, as it is influenced by a<br />

few drugs with a high budget impact. Many drugs are expected to have budget impacts<br />

well below that average.<br />

• Realistic scenario: average cost of € 2.135 million / drug / year<br />

• Low growth/cost scenario: average cost of € 2.0 million Euro / drug / year<br />

• High growth/cost scenario: average cost of € 2.3 million Euro / drug / year<br />

The chart below gives the results of the application of these scenarios, starting from a<br />

budget impact estimate of € 66 million <strong>for</strong> 2008. The SSF cost is not included. The<br />

application of the realistic scenario would lead to a budget impact of € 162 million in<br />

2013 or an increase of 145 % over 5 years. Although it is (also) difficult to estimate the<br />

total cost of drugs to the budget in five years, this amount should represent close to 2<br />

% of the total cost of drugs to the budget <strong>and</strong> over 10 % of the total drugs cost of<br />

hospitals.<br />

This growth <strong>for</strong>ecast is slower than the recent past, as based on an increase of 100 %<br />

between 2007 <strong>and</strong> 2008 (with an increase of 50 % in the number of reimbursed orphan<br />

drugs) <strong>and</strong> the estimated increase in the MORSE report 103 of the NIHDI was 50 %<br />

between 2007 <strong>and</strong> 2008 (that estimate covered only the 18 drugs with a college).<br />

The same restrictions that apply on the budget estimate <strong>for</strong> 2008 apply <strong>for</strong> the <strong>for</strong>ecast:<br />

• the basis <strong>for</strong> this <strong>for</strong>ecast is the estimate <strong>for</strong> 2008 which combines <strong>for</strong>ecasts<br />

<strong>and</strong> actual costs;<br />

• the parameters used <strong>for</strong> the <strong>for</strong>ecasts add to the uncertainty factor.


84 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

250<br />

200<br />

150<br />

100<br />

Figure 6.6 : Estimation of budget impact according to three scenario’s<br />

50<br />

0<br />

Key points<br />

2008 2009 2010 2011 2012 2013<br />

realistic<br />

• For 2008, the budget impact of orphan drugs in Belgium was estimated to<br />

range from 50 to 85 million Euro, which corresponds to over 5% of total<br />

hospital drugs budgets.<br />

• Three scenarios were applied to estimate the future budget impact: a<br />

conservative scenario (low growth/cost), a realistic scenario (best estimate)<br />

<strong>and</strong> a higher growth/cost scenario.<br />

• It was estimated that 10 new orphan drugs would reach market each year.<br />

• It was assumed that 90% of orphan drugs would gain reimbursement in<br />

Belgium.<br />

• It was estimated that the average cost of a reimbursed orphan drug would<br />

amount to 2.135 million Euro per year.<br />

• The realistic scenario would lead to a budget impact of orphan drugs of €<br />

162 million in 2013 or an increase of 145 % over 5 years. This would<br />

represent close to 4% of the cost of all drug reimbursements to the budget<br />

<strong>and</strong> over 10 % of total drugs costs of hospitals.<br />

Low<br />

High


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 85<br />

7 DISCUSSION AND CONCLUSIONS<br />

7.1 ORPHAN DRUG DESIGNATION AS A TACTICAL STEP<br />

One of the criticisms of the present system of <strong>Orphan</strong> Designation is that it allows<br />

medicinal products <strong>for</strong> ‘normal’ diseases to be designated as orphan drugs.<br />

This can happen when drugs are developed <strong>for</strong> a specific type of patients/disease (a<br />

practice called “targeting”), or when one disease is split into various sub-categories each<br />

presented with its own characteristics, a practice called “sub-setting” which is<br />

described above in the report (chapter 3):<br />

Sub-setting can lead to so-called “salami-slicing”: this is creating artificial subsets of a nonorphan<br />

condition, <strong>and</strong> basing the prevalence criterion on an unreal subpopulation. The aim is to<br />

obtain market exclusivity, a decrease of the costs <strong>and</strong> obligations linked to the registration<br />

dem<strong>and</strong>, <strong>and</strong> an increase of the exclusivity through new subpopulations (also known as the<br />

“evergreening tactic”).<br />

The industry is suspected of playing it tactically by introducing drugs to the market as<br />

‘orphan’, to fully reap the advantages (incentives) offered <strong>for</strong> the development of a drug<br />

with <strong>Orphan</strong> Designation, <strong>and</strong> then at a later stage to increase the number of<br />

indications <strong>for</strong> the same drug. This risk factor is increased by the fact that many<br />

oncological drugs obtain the orphan designation. As of today, one third of the orphan<br />

drugs on the Belgian market are <strong>for</strong> oncology, <strong>and</strong> their budget impact is also<br />

approximately one third of the budget impact of all orphan drugs.<br />

The COMP is very critical about the use of these techniques <strong>and</strong> adapts its own practice<br />

accordingly. Sub-setting is allowed under conditions. However, it seems impossible to<br />

exclude completely the possibility that manufacturers turn once orphan drugs into<br />

commercially highly profitable products later on.<br />

7.2 PREVALENCE VERSUS ECONOMIC MOTIVES<br />

As set out in Chapter three of this report, legislation on <strong>Orphan</strong> Designation at the EU<br />

level calls on two main criteria to decide on designation: either the (low) prevalence, or<br />

the high investment needed compared to the potential income.<br />

Both have the same underlying reasoning <strong>and</strong> are essentially considered to mean the<br />

same: a (very) low prevalence was <strong>for</strong> the legislator the equivalent to high investments<br />

<strong>for</strong> a potentially small market. The fact that both criteria are <strong>for</strong>mulated as “either / or”<br />

instead of “<strong>and</strong>” has however some consequences.<br />

Nearly all designations are granted based on prevalence. Only one designation was<br />

granted based on low ‘return on investment’: an <strong>Orphan</strong> Designation <strong>for</strong> a tropical<br />

neglected disease tt (tuberculosis) - not a rare disease. Only five dem<strong>and</strong>s were filed<br />

based on the return on investment criterion. With only one approval this means a very<br />

low success rate compared to the other criterion.<br />

Although judging the economic criteria is objectively speaking not particularly difficult, it<br />

faces a number of barriers: those who have to make the judgement have usually been<br />

trained in the field of health <strong>and</strong> do not have an economic background (hence lack the<br />

expertise) <strong>and</strong> the industry uses the argument that it is not possible to allocate costs<br />

clearly to one drug.<br />

This situation has become an issue that requires attention <strong>for</strong> the following reasons:<br />

• the high prices asked <strong>for</strong> orphan drugs raise the question to what extent<br />

these are indeed a fair reflection of the costs incurred by the industry –<br />

or rather just generate high profits <strong>for</strong> the industry;<br />

tt Interview with Dr. Jordi Llinares, EMEA, on the 14th of November 2008.


86 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

• it has been demonstrated uu that a few cases of orphan drugs which<br />

obviously required a very low level of investments have been brought to<br />

the market.<br />

Some orphan drugs generate revenues of hundreds of millions Euro per year. These are<br />

not yet “blockbusters” but these drugs obviously have reimbursed their initial<br />

investments <strong>and</strong> generate a high level of return to the sponsor. The legislator has<br />

<strong>for</strong>eseen the possibility to withdraw the market exclusivity after five years. This can be<br />

done at the initiative of an EU Member State. Yet, this has never happened up to now,<br />

<strong>and</strong> it seems unlikely that any individual Member State will take this initiative. The main<br />

reason why this is unlikely to happen is the absence of an agreement on what would be<br />

an acceptable return on investment.<br />

The overview below presents the trade-off between both interpretations of the<br />

legislation in terms of advantages <strong>and</strong> disadvantages.<br />

“either / or” “<strong>and</strong>”<br />

Advantages Stimulates innovation because less Would improve the application of<br />

barriers <strong>for</strong> industry<br />

the ‘spirit’ of the legislation<br />

Disadvantages Non-innovative, low investment Threshold <strong>for</strong> industry would be<br />

drugs can obtain orphan designation higher<br />

An adaptation of the legislation to “<strong>and</strong>” would potentially delay patient access to new<br />

drugs because two new barriers <strong>for</strong> the development of orphan drugs compared to the<br />

present situation would be created:<br />

• the need <strong>for</strong> industry to provide evidence (more work);<br />

• the need <strong>for</strong> COMP to evaluate based on economic criteria (need <strong>for</strong><br />

additional expertise).<br />

7.3 ASSESSING CLINICAL ADDED VALUE<br />

Assessing the clinical added value of orphan drugs is a challenge essentially because of<br />

the low number of patients. The techniques <strong>and</strong> st<strong>and</strong>ards used <strong>for</strong> drugs in general to<br />

confirm clinical effectiveness are difficult to apply on orphan drugs.<br />

At EMEA level, the clinical effectiveness is checked at the moment of deciding on<br />

Marketing Authorisation. Although the process is identical <strong>for</strong> orphan drugs compared<br />

to non-orphan drugs, there are guidelines that relate to clinical trials in small<br />

populations 32 .<br />

The clinical added value is assessed first by the CHMP as part of the Marketing<br />

Authorisation process <strong>and</strong> a second time at the national level <strong>for</strong> reimbursement. The<br />

same limited in<strong>for</strong>mation is used. The decision <strong>for</strong> market access is taken on absolute<br />

grounds (the drug is authorised to go to the market or not), the decision <strong>for</strong><br />

reimbursement on relative grounds (‘given the alternatives, this drug is worthwhile to<br />

be reimbursed”).<br />

At both decision levels, the analysis <strong>for</strong> orphans <strong>and</strong> <strong>for</strong> non-orphan drugs is done by<br />

the same organisations <strong>and</strong> based on the same criteria.<br />

Considering the clinical added value, most positive decisions taken by the CHMP to<br />

grant access to the market are based on a ‘benefit of the doubt’. For orphan drugs,<br />

there is seldom proof of clinical added value at that moment.<br />

The comparison that was per<strong>for</strong>med between the work undertaken at EU level <strong>and</strong> at<br />

national level in Belgium confirms that both analyses are based on nearly identical<br />

in<strong>for</strong>mation (see chapter 3 <strong>and</strong> 4).<br />

uu See the discussion of the ‘pricing issue’ in this chapter.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 87<br />

Even if the decisions to be taken on the basis of the same in<strong>for</strong>mation are different,<br />

there are obvious efficiency gains to be achieved.<br />

The EMEA decision process is more efficient compared to 27 individual national<br />

analyses <strong>for</strong> deciding on reimbursement. The work is actually done by two national<br />

Member States agencies, <strong>and</strong> a common opinion <strong>and</strong> decision is reached among all 27<br />

Member States at the CHMP. Creating a similar system specifically <strong>for</strong> the assessment of<br />

clinical added value <strong>and</strong> serving as input in the national decision regarding<br />

reimbursement at EU level would seem a logical next step.<br />

This has been suggested already at two occasions:<br />

• Eurordis (European Organisation <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong>) 104 made a<br />

recommendation in this respect. This recommendation is motivated by<br />

the differences in speed of market access among Member States. Bringing<br />

this aspect to the EU level would speed up decisions in Member States<br />

<strong>and</strong> avoid the present inequalities (industry concentrating on market<br />

access in procedurally easier or larger Member States);<br />

• The Pharmaceutical Forum 105 proposes an exchange of knowledge among<br />

Member States <strong>and</strong> to start an early dialogue between pricing <strong>and</strong><br />

reimbursement authorities.<br />

Another approach that is suggested (see chapter 2 above) is through the use of patient<br />

registries. An early patient registry, including data on the natural history of the rare<br />

disease <strong>and</strong> economically important variables, would allow regulatory authorities to<br />

follow up <strong>and</strong> evaluate long term continuous data collection <strong>and</strong> monitor the clinical<br />

efficacy over time. Setting up such patient registries is however a challenging task, as it<br />

would mean setting up a registry even be<strong>for</strong>e a drug is being developed. In practice it is<br />

uncertain <strong>for</strong> which rare disease a treatment will be developed. As the number of rare<br />

diseases is relatively high, questions about financing <strong>and</strong> governance of rare disease<br />

registries be<strong>for</strong>e the development of a treatment can be raised.<br />

An early patient registry, including data on the natural history of the disease <strong>and</strong><br />

economically relevant parameters, would allow regulatory authorities to follow up <strong>and</strong><br />

evaluate the uncertainties surrounding longer-term effectiveness <strong>and</strong> cost-effectiveness<br />

of an orphan drug in the relevant population. 25 Such an approach would support the<br />

decision-making process <strong>and</strong> allow more timely access to orphan drugs <strong>for</strong> patients. It<br />

would however not change the actual models on which decisions are based.<br />

The option to use disease <strong>and</strong> patient registries is described below.<br />

7.4 THE NEED FOR A RIGHT BALANCE BETWEEN ETHICAL<br />

AND ECONOMIC CONCERNS<br />

The development, marketing <strong>and</strong> reimbursement of orphan drugs challenge the general<br />

principles that underpin our current reimbursement policy.<br />

The average price of orphan drugs on the market today is high, which renders the<br />

current approach to orphan drugs potentially economically unsustainable. Moreover, it<br />

may be argued that it creates inequities because the life of one person is valued higher<br />

than the life of another. 22 This stretches the solidarity principle which underpins the<br />

health care system.<br />

The quote below from the conclusions of a NHS technology assessment study on<br />

Enzyme Replacement Therapy (ERT) <strong>for</strong> Fabry disease illustrates this situation:<br />

“Although ERT <strong>for</strong> treating the ‘average’ patient with Fabry’s disease exceeds the normal upper<br />

threshold <strong>for</strong> cost-effectiveness seen in NHS policy decisions by over sixfold, <strong>and</strong> the value <strong>for</strong><br />

MPS1 is likely to be of a similar order of magnitude, clinicians <strong>and</strong> the manufacturers argue<br />

that, as the disease is classified as an under European Union legislation, it has special status,<br />

<strong>and</strong> the NHS has no option but to provide ERT. More in<strong>for</strong>mation is required be<strong>for</strong>e the<br />

generalisability of the findings can be determined. Although data from the UK have been used<br />

wherever possible, this was very thin indeed.


88 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Nonetheless, even large errors in assumptions made will not reduce the ICER to anywhere near<br />

the upper level of treatments usually considered cost-effective.” 106<br />

The perceived extent of this problem is exacerbated by the fact that the costeffectiveness<br />

of orphan drugs is not assessed in Belgium at the moment of the<br />

reimbursement decision - as it is considered that the in<strong>for</strong>mation can never be<br />

sufficiently reliable due to the low number of patients.<br />

At the same time, fair distribution principles do not allow to exclude orphan drugs<br />

altogether from being reimbursed. Reimbursement of orphan drugs fit within the<br />

objectives of health care provision <strong>and</strong> fit within our system of social solidarity in which<br />

vulnerable groups receive support.<br />

And this may imply (limited) correction to market mechanisms. Furthermore, there is<br />

little support within the domain of social healthcare provision in general (in Belgium) <strong>for</strong><br />

the application of a pure cost-effectiveness <strong>and</strong> efficiency reasoning. 22<br />

The tension that currently exists between different societal concerns regarding orphan<br />

drugs needs to be addressed.<br />

The current situation leads to individual persons following their own ‘common sense’.<br />

Those who have to take decisions in the decision-making chain, from reimbursement<br />

decision <strong>for</strong> the drug to individual patient’s eligibility, are confronted with this dilemma<br />

<strong>and</strong> potential inequity. Anecdotal evidence collected shows that this ’tension‘ can lead<br />

to decisions like patients being refused a therapy because of age although this is<br />

nowhere mentioned as a criterion.<br />

A first step towards a solution to this situation would be to initiate a societal dialogue<br />

on the issue, to clarify what society wants <strong>and</strong> accepts in terms of ethical <strong>and</strong> economic<br />

consequences.<br />

7.5 PRICING<br />

From a regulatory point of view, the pricing of orphan drugs is not different to other<br />

drugs. The market conditions are however different to normal drugs.<br />

Facts <strong>and</strong> background:<br />

• The price is not an issue when the decision on market access is taken<br />

(EMEA – Marketing Authorisation).<br />

• The price is defined by the industry <strong>and</strong> submitted <strong>for</strong> approval to national<br />

authorities. In Belgium this is to the FPS Economy, in a process that runs<br />

in parallel to the reimbursement decision. The result is the acceptance of<br />

a maximum price. The analysis per<strong>for</strong>med by the FPS Economy is mainly<br />

based on comparisons. For orphan drugs, this means comparison with<br />

other countries.<br />

• Price negotiations are in principle not part of the drug reimbursement<br />

decision process. The price approved by the FPS Economy is considered<br />

to be the basis <strong>for</strong> reimbursement.<br />

• There is generally no negotiation on the price. The only negotiation that<br />

may occur is by the government, between the advice of the DRC <strong>and</strong> the<br />

actual (publication of the) decision. This is mainly linked to the budget<br />

impact <strong>and</strong> can lead to a compromise with the industry to get approval.<br />

Price could be an element in the negotiation, but in practice it is not.<br />

• The DRC could (re-)negotiate the price at regular revisions. This has<br />

however not yet happened <strong>for</strong> orphan drugs.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 89<br />

Small monopolies<br />

The price is an essential element of the context created by the orphan drug legislation.<br />

<strong>Drugs</strong> to treat rare diseases are considered as a small market, which has a number of<br />

implications:<br />

the investment <strong>for</strong> industry is high compared to the potential market size;<br />

the risks <strong>for</strong> the industry are high in terms of return on their investment.<br />

Legitimate concerns that these factors would discourage industry from developing<br />

orphan drugs have led to legislation which aims to reduce the risks, providing incentives<br />

<strong>for</strong> investments in research <strong>for</strong> rare diseases <strong>and</strong> <strong>for</strong> the market introduction of orphan<br />

drugs. This legislation has created a com<strong>for</strong>table situation <strong>for</strong> the pharmaceutical<br />

industry:<br />

they obtain market exclusivity <strong>for</strong> orphan drugs (no direct competition);<br />

the price is set by the industry <strong>and</strong> is not negotiated by any party.<br />

The end result is that small ‘virtual monopolies’ are created in which the industry is free<br />

to ask the price they want <strong>for</strong> orphan drugs. These often high prices are justified by the<br />

need to reimburse the research <strong>and</strong> development costs.<br />

There are however no market mechanisms in place to correct a potentially too high<br />

price:<br />

• there is no direct or indirect competition, as is the case <strong>for</strong> other drugs;<br />

• customers have no bargaining power towards the industry;<br />

• the market is closed <strong>for</strong> competition: no competitor will run the risk to<br />

invest in an alternative medicinal product as the legislation blocks the<br />

(small) market access <strong>for</strong> ten years.<br />

As such, the legislation which has a favourable impact in terms of the supply of orphan<br />

drugs to the market has, through its distortion of the market mechanisms, also the<br />

adverse effect of too high prices not being adjusted. The current rules do not, however,<br />

preclude the production of generics or biosimilars <strong>for</strong> orphan drugs once the period of<br />

market exclusivity is passed. The production of generics or biosimilars may in the<br />

medium term reduce the prices of orphan drugs.<br />

Adding to the problem is the fact that the market <strong>for</strong> drugs in general <strong>and</strong> <strong>for</strong> orphans,<br />

is not transparent. In<strong>for</strong>mation on effectiveness <strong>and</strong> hence cost-effectiveness of the<br />

treatment is not available at the moment of market access, <strong>and</strong> is not available post<br />

Marketing Authorisation either. In<strong>for</strong>mation on clinical effectiveness is frequently (very)<br />

limited, as explained above, <strong>and</strong> decisions to grant market access are often taken<br />

allowing the ‘benefit of the doubt’.<br />

“Identical” medicines<br />

Three of the 48 orphan drugs with Marketing Authorisation at the European level are<br />

drugs that have a “twin” product on the market. These twins are <strong>for</strong> other, non orphan<br />

indications <strong>and</strong> have a different br<strong>and</strong> name.<br />

The orphan version of the twin is always marketed at a higher price.<br />

The best known example is Revatio® which is another name <strong>for</strong> Viagra®. The two<br />

other products are:<br />

• Savene® (orphan) being the same product as Cardioxane®. Savene® is<br />

sold in Belgium, Cardioxane® not. If it was available on the Belgian<br />

market, one can assume it would be prescribed instead of Savene®<br />

• Siklos® (orphan) is identical to Hydrea®: one is delivered as capsules, the<br />

other in the <strong>for</strong>m of tablets. Siklos® is not on the Belgian market <strong>and</strong> one<br />

can assume that patients of well in<strong>for</strong>med medical doctors are receiving<br />

Hydrea®.


90 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

In the example of Revatio® / Viagra®, both produced by the same company, the<br />

decision to develop a different br<strong>and</strong> <strong>for</strong> the orphan indication is almost certainly not<br />

motivated by a return on investment need. Rather, the orphan drug legislation provides<br />

an additional incentive <strong>for</strong> the industry to explore <strong>and</strong> introduce the drug in a specific<br />

market niche. Since investments are largely covered by the profits generated by Viagra,<br />

the ‘additional cost’ is essentially linked to clinical trials <strong>and</strong> to marketing.<br />

Compounding preparations<br />

Two orphan drugs that obtained a market authorisation from EMEA were refused<br />

reimbursement in Belgium because of the existence of an alternative in the <strong>for</strong>m of a<br />

compounding preparation.<br />

Although both cannot be compared as one is an artisanal product <strong>and</strong> the other an<br />

industrial product, the price difference was such that it prevented approval.<br />

Conclusion<br />

The pricing issue is a key element of the equation. The orphan drug legislation creates a<br />

positive market environment through incentives, one of which is the creation of small<br />

virtual monopolies. Industry behaviour is to ask <strong>for</strong> high prices. Member States have<br />

little negotiation power <strong>and</strong> there are no market mechanisms in place to put a<br />

downward pressure on prices.<br />

The spirit of the legislation, being to stimulate research <strong>and</strong> development on drugs <strong>for</strong><br />

diseases that would otherwise be neglected by industry <strong>and</strong> academia, is put at risk by<br />

this situation, as high prices also mean high budget impacts <strong>and</strong> in general low costeffectiveness<br />

in comparison to non-orphan drugs.<br />

Three potential routes to solve the existing problem are:<br />

• an adaptation of the legislation to ensure that its application happens<br />

more according to its spirit with analysis of the return on investment<br />

including subsidies received <strong>for</strong> R&D <strong>and</strong> justification of price setting;<br />

• the application of risk-sharing systems like price-<strong>for</strong>-per<strong>for</strong>mance schemes<br />

or conditional reimbursements<br />

• the organisation of price negotiations at the EU-level instead of at<br />

Member State level, which could be combined with both previous bullet<br />

points.<br />

Advantages <strong>and</strong> disadvantages of the first two are:<br />

In<strong>for</strong>mation on return on investment Risk sharing<br />

advantages • Is a logical consequence of the<br />

existing legislation, which should<br />

there<strong>for</strong>e be acceptable to all<br />

stakeholders<br />

disadvantages • This in<strong>for</strong>mation needs to be<br />

assessed by experts<br />

• No need to provide in<strong>for</strong>mation<br />

on investments <strong>and</strong> potential<br />

returns<br />

• Need to define per<strong>for</strong>mance<br />

criteria<br />

• Is a new technique, there is little<br />

experience with this type of pricesetting<br />

(in Belgium)


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 91<br />

7.6 EXTENSION OF INDICATIONS<br />

Designation <strong>for</strong> an orphan drug is possible <strong>for</strong> indications with a prevalence up to 5 in<br />

10,000.<br />

In practice most orphan drugs are <strong>for</strong> ultra rare diseases, e.g.:<br />

Disease Prevalence <strong>Orphan</strong> drug<br />

Fabry disease 1.75 / 100,000 Fabrazyme® & Replagal®<br />

MPS I 1.3 / 100,000 Aldurazyme®<br />

MPS II 0.6 / 100,000 Elaprase®<br />

MPS IV 0.4 / 100,000 Naglazyme®<br />

Acute promyelocyctic leukemia 8 / 100,000 Trisenox®<br />

Chronic myeloid leukemia 6 / 100,000 Glivec®<br />

Cases exist where a drug obtained the designation <strong>and</strong> Marketing Authorisation <strong>for</strong> one<br />

indication, <strong>and</strong> then later this is extended to more indications.<br />

The legislation allows this. The same product can have more indications, <strong>and</strong> the<br />

prevalences <strong>for</strong> the various indications are not “added up”.<br />

<strong>Orphan</strong> drugs with more than one orphan indication at European <strong>and</strong> Belgian level are:<br />

Drug Number of indications EMEA Number of indications Belgium<br />

Glivec® 6 2<br />

Nexavar® 2 2<br />

Sprycel® 2 1<br />

Sutent® vv 2 2<br />

Tracleer® 2 2<br />

Changing the legislation to link the designation as orphan drug to the total prevalence of<br />

all indications would have consequences as described in the table below:<br />

Advantage Disadvantage<br />

Change the legislation • Would be more in line<br />

with spirit of legislation<br />

• Would ensure to<br />

concentrate on really<br />

rare<br />

• Would create a barrier<br />

to use the OD<br />

legislation <strong>for</strong> purposes<br />

it was not meant <strong>for</strong><br />

vv Sutent® has withdrawn its orphan designation at EMEA level<br />

• Creates a<br />

potential barrier<br />

to develop new<br />

product-indication<br />

combinations


92 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

7.7 GROWTH OF THE BUDGET IMPACT OF ORPHAN DRUGS<br />

Total budgets <strong>for</strong> orphan drugs were very small when the legislation was launched in<br />

2000.<br />

Today they have become significant, even if the total number of patients treated is still<br />

limited.<br />

This high growth is powered by a number of factors:<br />

• the high average price of orphan drugs;<br />

• the steady increase in the number of orphan drugs coming to the market.<br />

In Belgium, there is no budget ceiling <strong>for</strong> orphan drugs, but the total cost of all drugs<br />

reimbursed does have a ceiling. When the global ceiling is reached, there are<br />

mechanisms to compensate <strong>for</strong> over-expenditure. The government can charge an<br />

alternative charge to the pharmaceutical industry to compensate 100% of the overexpenditure,<br />

with a maximum of €100 million per year. <strong>Orphan</strong> drugs do not contribute<br />

to this subsidiary charge. <strong>Orphan</strong> drugs as a group have no ceiling.<br />

The budget increase of orphan drugs there<strong>for</strong>e puts pressure on the total ceiling, <strong>and</strong><br />

non orphan drugs industry is likely to pay <strong>for</strong> the orphan drugs industry.<br />

The cost of orphan drugs in Belgium is estimated to have been over 5 % of total hospital<br />

drug budgets in 2008 ww <strong>and</strong> further estimates indicate the future cost could be well<br />

above 10 % of hospital drug budgets in five years from now. <strong>Orphan</strong> drugs represent<br />

probably 2% of total drug reimbursement costs in 2009, <strong>and</strong> could represent close to<br />

4% in 2013.<br />

This high cost creates an upward pressure on health insurance budgets. If <strong>and</strong> when<br />

hundreds of orphan drugs become available, they would still cover only part of the<br />

needs of all the patients suffering from rare diseases. Moreover, relatively large amounts<br />

of the limited health care budget would go to a few patients, which may challenge the<br />

boundaries of solidarity. Based on the experience with a first set of 31 orphan drugs,<br />

the cost to the health insurance system under the present conditions could become<br />

unbearable.<br />

The high prices combined with the growing budget impact of orphan drugs also<br />

negatively affect the image of the orphan drugs among decision-makers. Globally<br />

speaking, the orphan drug legislation is considered by all parties to be a success. The<br />

price of this success is the rising budget. The negative image created puts the success at<br />

risk.<br />

7.8 VARIATIONS IN ACCESS AND USE AMONG MEMBER<br />

STATES<br />

Although the Marketing Authorisation decision grants access to the market in 27<br />

Member States, because of the cost of the drugs, effective access is reached only when<br />

the decision is taken to reimburse the medicinal product (at the national level).<br />

As a consequence, the effective market access <strong>and</strong> the utilization of orphan drugs vary<br />

among Member States.<br />

The situation of Belgium compared to other countries analysed can be summarised as<br />

follows:<br />

• Starting a process to decide on reimbursement of a drug is the initiative of<br />

industry. In practice, Belgium is not one of the countries that is chosen as<br />

a priority by industry.<br />

ww The number of individual patients treated is difficult to estimate both <strong>for</strong> orphan drug treatments <strong>and</strong> <strong>for</strong><br />

hospital treatments, but one speaks of a difference in cost/treatment/patient that must be in the around<br />

one (<strong>for</strong> non orphan drugs) to one thous<strong>and</strong> (<strong>for</strong> orphan drugs).


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 93<br />

• Out of the 47 orphan drugs having obtained Marketing Authorisation by<br />

end 2008, 31 are reimbursed in Belgium <strong>and</strong> 4 are not reimbursed. This<br />

puts Belgium at third place compared to France, Italy, the Netherl<strong>and</strong>s<br />

<strong>and</strong> Sweden (see point 4.9). xx<br />

• The procedure to access reimbursement in Belgium takes in theory 180<br />

days, but will in practice, due to interruptions, last longer. There is little<br />

evidence that this process ends up to be much longer than in other<br />

Member States, but industry mentions this as the argument to be reticent<br />

to start reimbursement procedure. In comparison, an orphan drug having<br />

obtained Marketing Authorisation is automatically launched on the British<br />

market. But if the advice of NICE is requested <strong>for</strong> reimbursement, the<br />

procedure can be slow (see point 4.8).<br />

• In Belgium three systems exist <strong>for</strong> early access: the SSF, the medical needs<br />

programme <strong>and</strong> the compassionate use legislation (see 4.3.3.2 <strong>and</strong><br />

4.3.3.3). The SSF de facto plays a role between Marketing Authorisation<br />

<strong>and</strong> reimbursement decision. It can be questioned if the SSF is an<br />

adequate mechanism <strong>for</strong> that purpose (awareness of patients <strong>and</strong> Medical<br />

Doctors, criteria, etc.).<br />

Early access (be<strong>for</strong>e Marketing Authorisation)<br />

In Belgium, early access is possible through the Special Solidarity Fund (see point<br />

4.3.3.3).<br />

Figure 7.1 : SSF reimbursement <strong>for</strong> <strong>Orphan</strong> <strong>Drugs</strong> with MA in 2007<br />

<strong>Orphan</strong> drug Total NIHDI Number of patients NIHDI expenditures<br />

Expenditures<br />

per patient<br />

Myozyme® € 3,540,723 7 € 505,818<br />

Revatio® € 299,358 € 67 € 4,468<br />

Revlimid® € 141,050 € 57 € 2,475<br />

Ventavis/Iloprost® € 100,927 € 9 € 11.214<br />

Tracleer® € 2,167 € 1 € 2,167<br />

Source: NIHDI. Jaarverslag 2007 betreffende het Bijzonder Solidariteitsfonds, 2008<br />

Several Member States have a particular procedure providing early access:<br />

• In France, orphan drugs can be accessed be<strong>for</strong>e they are reimbursed,<br />

through the ‘Authorisation <strong>for</strong> Temporary Use’ (ATU) procedure (see<br />

point 4.4).<br />

• A special fund <strong>for</strong> compassionate use was set up in Italy in order to<br />

finance early access to orphan drugs <strong>and</strong> trials on rare diseases (see point<br />

4.5).<br />

xx In<strong>for</strong>mation about the United Kingdom is not available.


94 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

7.9 AWARENESS RAISING<br />

Because of the low incidence of rare diseases, health professionals generally have a low<br />

awareness <strong>and</strong> little knowledge on how to treat such diseases.<br />

This explains the importance <strong>and</strong> focus of the EU level actions <strong>and</strong> national plans <strong>for</strong><br />

rare diseases on:<br />

• developing awareness raising actions <strong>and</strong> tools;<br />

• developing tools to give access to knowledge <strong>and</strong> expertise including databases;<br />

• building expertise in specialized centres; <strong>for</strong> many Member States this<br />

implies to refer patients with a specific disease to a specific expert centre;<br />

• the added value of EU <strong>and</strong> international cooperation.<br />

The concept of “orphan drug” is a non-reality <strong>for</strong> the medical professionals. Their<br />

concern is the patient, the disease <strong>and</strong> the potential treatment. Whether the drug is an<br />

orphan drug or not has no importance. “<strong>Orphan</strong> drugs” is a technical concept, not<br />

understood, not commonly used by health professionals.<br />

Even <strong>for</strong> specialists on rare diseases <strong>and</strong> orphan drugs, there exists confusion on which<br />

drugs are “orphan” <strong>and</strong> which are not. Indeed, there exists a ‘grey zone’:<br />

• drugs <strong>for</strong> orphan indications that do not have the orphan status, either<br />

because they were introduced be<strong>for</strong>e the legislation, or because they are<br />

also used <strong>for</strong> non-orphan indications; some of these drugs do have the<br />

orphan status in the USA but (not yet) at EU level;<br />

• there is one drug that has the orphan status in Belgium but not at EU<br />

level yy ;<br />

• there are various drugs with orphan status at EU level or in the US, that<br />

are not reimbursed in Belgium;<br />

• etc.<br />

The only efficient solution to close this awareness gap seems to be to concentrate<br />

expertise. The route of setting up expert centres <strong>for</strong> rare diseases will also be beneficial<br />

<strong>for</strong> the promotion of therapies using the right orphan drug.<br />

7.10 COLLEGES AND CONTROL OF ELIGIBILITY<br />

In Belgium, orphan drugs can only be prescribed to individual patients if a number of<br />

conditions are met. These are defined by the DRC at the moment of the decision to put<br />

the drug on the list of reimbursed orphan drugs. Medical Doctors who wish to<br />

prescribe the drug have to ensure these conditions are met <strong>and</strong> confirm this when<br />

sending the application <strong>for</strong> the reimbursement to the health insurance organisations.<br />

In principle, this is linked to the control of the eligibility. It also serves to collect<br />

in<strong>for</strong>mation that could be used later <strong>for</strong> revisions of decisions.<br />

In practice, the perception by Medical Doctors <strong>and</strong> the industry is that criteria <strong>and</strong><br />

conditions (of which the relevance is not always clear) are added as a technique to<br />

create barriers to the use of orphan drugs. Examples are the need to renew the<br />

dem<strong>and</strong> every six or twelve months <strong>for</strong> life-time diseases, or to repeat tests at regular<br />

intervals, which leads to (unnecessary) costs <strong>and</strong> a sometimes heavy physical <strong>and</strong> mental<br />

burden <strong>for</strong> the patient. This perception seems to be exacerbated by a lack of<br />

transparency <strong>and</strong> of return on the in<strong>for</strong>mation provided.<br />

yy Duodopa®, which obtained the designation at national level in 2006; this would not be possible anymore<br />

today.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 95<br />

The Colleges of <strong>Orphan</strong> <strong>Drugs</strong>, when they exist, have the power to adapt <strong>and</strong> change<br />

these conditions, based on experience. This is apparently working, but is not done<br />

systematically or pro-actively although the Colleges do have a <strong>for</strong>mal responsibility in<br />

this respect. It does put an additional burden <strong>and</strong> responsibility on the Colleges.<br />

The following observations can be made in relation to the functioning <strong>and</strong> the role of<br />

the Colleges:<br />

• the establishment of Colleges seems to be a good technique, as it allows<br />

to bring together the (rare) expertise;<br />

• the sickness funds are not legally required to ask the advice of the<br />

Colleges, but they nevertheless have a consensus to request it<br />

systematically;<br />

• the work volumes <strong>for</strong> these Colleges is very high, <strong>and</strong> the increase in the<br />

number of orphan drugs leads to the need to create a permanent support<br />

structure;<br />

• Colleges are reactive in their functioning; they do have a permanent<br />

structure but with little resources. With more resources, they could<br />

become more pro-active <strong>and</strong> propose changes <strong>and</strong> improvements;<br />

• their success raises the question as to what to do with the thirteen drugs<br />

out of 31 that have no College: should this not be systematic? ;<br />

• if a sickness fund takes a negative decision, it has the theoretical obligation<br />

to in<strong>for</strong>m the College but there is little evidence that this leads to<br />

additional knowledge. If the Colleges reviewed all requests <strong>and</strong> registered<br />

systematically all decisions, both positive <strong>and</strong> negative, they would be in a<br />

better position to provide advice <strong>and</strong> in<strong>for</strong>mation at the moment of<br />

revisions;<br />

• Colleges potentially pool a lot of in<strong>for</strong>mation; it would be easy to ensure<br />

they concentrate all in<strong>for</strong>mation. For the moment, the collected<br />

in<strong>for</strong>mation is not easily available, e.g. the year reports are not publicly<br />

available. Making the in<strong>for</strong>mation publicly available would improve the<br />

efficiency, also to decide on revisions.<br />

Typology of the Colleges:<br />

Total number of Colleges 18<br />

Therapeutic area Endocrinology/Metabolism: 10<br />

Neurology: 1<br />

Cardiovascular/respiratory: 3<br />

Oncology: 3<br />

Haematology: 1<br />

First or second line 11 First<br />

1 First/second<br />

6 Second<br />

Alternative? 9 have an alternative versus 9<br />

Treatment 8 peroral (by mouth)<br />

10 paranteral


96 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

7.11 USE OF REGISTRIES<br />

Patient or disease registries zz are used <strong>for</strong> various purposes <strong>and</strong> particularly in the case<br />

of rare diseases. Setting up patient <strong>and</strong> disease registries is part of the EU policy <strong>and</strong> is<br />

an action line in national rare disease plans that many Member States have or are setting<br />

up.<br />

Typical purposes why registries are being set up are:<br />

• to describe the natural history of a disease;<br />

• <strong>for</strong> research purposes (e.g. to have fast access to patients);<br />

• to determine clinical effectiveness;<br />

• to monitor cost-effectiveness;<br />

• to monitor safety <strong>and</strong> harm.<br />

Registries are set up <strong>for</strong> rare diseases be<strong>for</strong>e <strong>and</strong> independently of the fact a medicinal<br />

product is being developed. As there are an estimated 5,000 to 8,000 rare diseases, it is<br />

clear that registries exist <strong>for</strong> only part of the rare diseases.<br />

With regard to orphan drugs, there are two moments when registries are usually set<br />

up:<br />

• at the moment of Marketing Authorisation. The CHMP will on an ad hoc<br />

basis impose on the industry to set up a registry. This can be <strong>for</strong> various<br />

purposes mainly linked to the clinical effectiveness <strong>and</strong>/or the safety <strong>and</strong><br />

harm monitoring;<br />

• at the moment of the decision on reimbursement, national authorities can<br />

also decide that setting up a registry is a condition <strong>for</strong> reimbursement.<br />

Each of these decisions is ad hoc, there is no st<strong>and</strong>ardisation, neither at EMEA level, nor<br />

at the level of individual Member States, nor among Member States.<br />

An exception is the MPS registry in the UK, that was set up be<strong>for</strong>e a drug <strong>for</strong> MPS was<br />

developed.<br />

The industry is in charge of funding <strong>and</strong> setting up these registries which goes against<br />

the important principle that data should be “independent” in the case of registries. They<br />

do this adequately but can decide autonomously on how <strong>and</strong> according to which<br />

st<strong>and</strong>ards it will be set up <strong>and</strong> managed nearly always by third parties.<br />

The existence of registries <strong>for</strong> the indications of orphan drugs is generally considered as<br />

an advantage offsetting the potential disadvantages (e.g. cost or privacy issues) <strong>and</strong> the<br />

difficulties linked to their management particularly to ensure their long term<br />

sustainability beyond the point in time where obligations <strong>for</strong> the EMEA are fulfilled. The<br />

main advantages are:<br />

• access to patients: both <strong>for</strong> research <strong>and</strong> market access (linked to the<br />

rarity);<br />

• transparency: it is a source of in<strong>for</strong>mation <strong>for</strong> those who need to decide<br />

on the most adequate therapy (effective treatment);<br />

• control: it is a way <strong>for</strong> the reimbursement authorities to control whether<br />

the medicine is prescribed <strong>for</strong> the right type of patients, as well as to gain<br />

insights on whether the therapy is working <strong>and</strong> should be continued.<br />

zz Disease registry is a specially designed database with voluntary, observational clinical data collected<br />

from physicians <strong>and</strong> intended to explore <strong>and</strong> define the natural course <strong>and</strong> clinical characteristics of<br />

disease, as well as to track <strong>and</strong> characterize response to treatment.<br />

Patient register is a database (list) containing baseline in<strong>for</strong>mation on the existence of patients with (a)<br />

certain disease(s), but without any longitudinal follow-up.<br />

(Working Group Pricing <strong>and</strong> Reimbursement. Improving access to orphan medicines <strong>for</strong> all affected EU<br />

citizens. The Pharmaceutical Forum. 2008. Available from<br />

[Last accessed: 10/12/2008].)


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 97<br />

Willingness to participate in patient <strong>and</strong> disease registries is high among the medical<br />

professionals, which contrasts with their reluctance to provide a lot of in<strong>for</strong>mation<br />

linked to the decisions <strong>for</strong> individual reimbursement. This difference is explained by the<br />

return <strong>for</strong> the Medical Doctor, which is real with a registry <strong>and</strong> most often absent <strong>for</strong><br />

the latter.<br />

Transparency is an important value in the case of orphan drugs. Decisions on market<br />

authorisation are based on limited clinical evidence. Reimbursement decisions are based<br />

on the same limited clinical evidence, but are furthermore taken without in<strong>for</strong>mation on<br />

cost-effectiveness. When the medicinal product is on the market, the normal market<br />

mechanisms are not working as there is no alternative to the treatment which is<br />

rein<strong>for</strong>ced by the orphan drug legislation (market exclusivity). The transparency<br />

achieved through registries can compensate <strong>for</strong> this, especially by providing growing<br />

evidence on both the clinical effectiveness <strong>and</strong> the cost-effectiveness of the treatment. aaa<br />

Various routes exist to improve the use of registries in the case of orphan drugs:<br />

1. systematically setting up patient registries <strong>for</strong> all indications <strong>for</strong> which<br />

designations were granted; this will create value <strong>for</strong> all those involved later in<br />

the decision-making, including industry who will have easier access to<br />

patients, <strong>and</strong> <strong>for</strong> reimbursement authorities who can <strong>for</strong>ecast the budget<br />

impact more precisely;<br />

2. st<strong>and</strong>ardizing the registries, at the EU level <strong>for</strong> clinical evidence;<br />

3. ensure coordination <strong>for</strong> aspects of clinical evidence between what is asked at<br />

EU level <strong>and</strong> what is asked at national level;<br />

4. systematically set up data collection on cost-effectiveness of treatments<br />

through registries;<br />

5. coordinate the cost-effectiveness in<strong>for</strong>mation to be collected between<br />

Member States.<br />

This subject is worth a study on its own, but it is clear that value can be created<br />

through:<br />

• st<strong>and</strong>ardization <strong>and</strong> coordination between the various decision-makers;<br />

• collecting in<strong>for</strong>mation on the effectiveness of the treatments / drugs after<br />

their introduction on the market. This will improve the quality of<br />

in<strong>for</strong>mation available when revising reimbursement decisions;<br />

• transparency: it will allow the “market” to function better by ensuring the<br />

in<strong>for</strong>mation flows.<br />

aaa An analysis based on the registry has however no added value <strong>for</strong> the assessment of the “incremental”<br />

cost-effectiveness of that respective drug, as such assessment requires a comparison with the “best<br />

alternative treatment”. Cost benefit can only be compared if data on the alternative treatment is available.<br />

A registry is (only) a way to monitor the effectiveness of a medicinal product.


98 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Key points<br />

• The present system of <strong>Orphan</strong> Designation allows <strong>for</strong> medicinal products <strong>for</strong><br />

‘normal’ diseases to be designated as orphan drugs.<br />

• The economic factors underlying <strong>Orphan</strong> Designation can be questioned in<br />

some cases as a low prevalence does not equal potential low return on<br />

investment.<br />

• Evidence about clinical added value of orphan drugs is rarely available at the<br />

moment of registration due to the low number of patients. European<br />

cooperation can lead to significant efficiency gains, particularly through the<br />

use of patient registries.<br />

• There is a need to find a right balance between ethical <strong>and</strong> economic<br />

concerns as this leads to tensions. A solution could be to initiate a societal<br />

dialogue on the issue, to clarify what society wants <strong>and</strong> accepts in terms of<br />

ethical <strong>and</strong> economic consequences<br />

• In essence, small monopolies are created to stimulate development <strong>and</strong><br />

supply of orphan drugs, but there are no market mechanisms to adjust<br />

prices once drugs are on the market.<br />

• The growing impact of orphan drugs on the total budget <strong>for</strong> health<br />

insurance in Belgium is creating pressures.<br />

• Indications can be extended <strong>for</strong> an orphan drug <strong>and</strong> the total prevalence<br />

across indications is not considered.<br />

• Access of Belgian patients to orphan drugs in practice depends on the<br />

manufacturer submitting a reimbursement application <strong>and</strong> on the Drug<br />

Reimbursement Committee granting reimbursement.<br />

• Health professionals generally have a low awareness of rare diseases <strong>and</strong><br />

orphan drugs due to their rare occurrence.<br />

• There is a need <strong>for</strong> a comprehensive approach towards defining <strong>and</strong><br />

exercising the role of the Colleges of <strong>Orphan</strong> <strong>Drugs</strong>.<br />

• There is a need <strong>for</strong> a European st<strong>and</strong>ardised approach to setting up <strong>and</strong><br />

using patient <strong>and</strong> disease registries. Centralisation will also increase the<br />

chances of long term sustainability.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 99<br />

8 APPENDICES


100 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

8.1 OVERVIEW REIMBURSED ORPHAN DRUGS IN BELGIUM<br />

Drug Indication Sponsor EMEA NIHDI<br />

<strong>Orphan</strong><br />

Designation<br />

MA<br />

Submission<br />

date<br />

Approval<br />

date<br />

Reimbursed<br />

since<br />

Aldurazyme<br />

®<br />

Treatment of Mucopolysaccharidosis, type I Genzyme 14/2/2001 10/6/2003 26/6/2003 20/7/2004 1/8/2004<br />

Atriance® Acute lymphoblastic leukaemia GSK 16/6/2005 22/8/2007 19/9/2007 21/5/2008 1/6/2008<br />

Busilvex® Hematopoietic cell transplantation Pierre Fabre Medicament 29/12/2000 9/7/2003 7/6/2004 13/12/2004 1/10/2008<br />

Carbaglu® NAGS deficiency <strong>Orphan</strong> Europe 18/10/2000 24/1/2003 7/12/2005 21/8/2006 1/9/2006<br />

Duodopa® Parkinson Solvay / / 7/7/2006 16/2/2007 1/3/2007<br />

Elaprase®<br />

Mucopolysaccharidosis, type II (Hunter<br />

Syndrome)<br />

Shire 11/12/2001 8/1/2007 13/3/2007 20/12/2007 1/1/2008<br />

Evoltra® Acute lymphoblastic leukaemia Genzyme 5/2/2002 29/5/2006 7/1/2008 20/6/2008 1/7/2008<br />

Exjade®<br />

Chronic iron overload requiring chelation<br />

therapy<br />

Novartis Pharma 13/3/2002 28/8/2006 6/11/2006 20/6/2007 1/8/2007<br />

Fabrazyme® Fabry disease Genzyme 8/8/2000 4/5/2001 8/4/2002 20/7/2004 1/8/2004<br />

Glivec®<br />

Increlex®<br />

Chronic myeloid leukaemia<br />

7/7/2003 19/3/2004<br />

Novartis 14/2/2001 27/8/2001<br />

Malignant gastrointestinal stromal tumours 5/8/2002 20/6/2003<br />

Treatment of primary insulin-like growth factor-1<br />

deficiency due to molecular or genetic defects<br />

(primary growth hormone insensitivity syndrome)<br />

1/7/2003<br />

Ipsen 22/5/2006 3/8/2007 3/12/2007 18/7/2008 1/8/2008<br />

Lysodren® Adrenal cortical carcinoma HRA Pharma 12/6/2002 28/4/2004 21/11/2006 20/12/2007 1/1/2008<br />

Myozyme®<br />

Naglazyme®<br />

Glycogen Storage Disease type II (Pompe´s<br />

disease)<br />

Treatment of Mucopolysaccharidosis, type VI<br />

(Maroteaux-Lamy Syndrome)<br />

Genzyme 14/2/2001 29/3/2006 18/5/2006 20/4/2007 1/5/2007<br />

Biomarin Europe 14/2/2001 24/1/2006 21/11/2007 20/11/2008 1/12/2008


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 101<br />

Drug Indication Sponsor EMEA NIHDI<br />

Nexavar®<br />

<strong>Orphan</strong><br />

Designation<br />

MA<br />

Submission<br />

date<br />

Approval<br />

date<br />

Reimbursed<br />

since<br />

Renal cell carcinoma<br />

1/8/2006 21/3/2007 1/4/2007<br />

Bayer Healtcare 29/7/2004 19/7/2006<br />

Hepatocellular carcinoma 2/10/2007 20/6/2008 1/7/2008<br />

Orfadin® Tyrosinaemia type 1 Swedish <strong>Orphan</strong> 29/12/2000 21/2/2005 3/8/2005 20/6/2006 1/7/2006<br />

Replagal® Fabry disease TKT-Europe / Shire 8/8/2000 4/5/2004 Unknown Unknown<br />

Revatio® Pulmonary Arterial Hypertension Pfizer 12/12/2003 28/10/2005 24/5/2006 21/5/2007 1/6/2007<br />

Revlimid® Multiple Myelome Celgene 12/12/2003 14/6/2007 19/7/2007 21/3/2008 1/4/2008<br />

Savene® Anthracycline extravasations Topotarget 10/9/2001 28/7/2006 14/12/2006 21/8/2007 1/9/2007<br />

Somavert® Acromegaly Pfizer 14/2/2001 13/11/2002 27/6/2003 19/3/2004 1/4/2004<br />

Sprycel® Chronic myeloid leukaemia Bristol-Myers Squibb 23/12/2005 20/11/2006 14/12/2006 21/8/2007 1/9/2007<br />

Sutent®<br />

1/8/2004<br />

Malignant gastrointestinal stromal tumours<br />

9/8/2006 21/3/2007 1/4/2007<br />

Pfizer<br />

Renal cell carcinoma 8/2/2007 20/7/2007 1/8/2007<br />

Tasigna® Chronic myeloid leukaemia Novartis Pharma 13/4/2007 19/11/2007 17/12/2007 21/8/2008 1/9/2008<br />

Thalidomide<br />

®<br />

Multiple Myelome Celgene 20/11/2001 16/4/2008 Unknown Unknown 18/09/1999<br />

Thelin® Pulmonary Arterial Hypertension Pfizer / Encysive UK Ltd 21/10/2004 10/8/2006 19/4/2007 20/12/2007 1/1/2008<br />

Torisel® Renal cell carcinoma Wyeth 6/4/2006 19/11/2007 4/1/2008 20/11/2008 1/12/2008<br />

Tracleer®<br />

Pulmonary Arterial Hypertension<br />

Chronic thromboembolic pulmonary<br />

hypertension<br />

Actelion 14/2/2001 15/5/2002<br />

8/1/2003 20/7/2004 1/8/2004<br />

21/12/2005 21/8/2006 1/9/2006<br />

Trisenox® Acute promyelocytic leukaemia Cephalon 18/10/2000 5/3/2002 16/2/2005 20/10/2005 1/11/2005<br />

Xagrid® Essential thrombocytose Shire 29/12/2000 16/11/2004 24/1/2005 20/10/2005 1/11/2005<br />

Zavesca® Gaucher disease Actelion 18/10/2000 20/11/2002 30/11/2004 19/8/2005 1/9/2005


102 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

8.2 CHARACTERISTICS OF ORPHAN DRUGS SUBMITTED FOR REIMBURSEMENT IN BELGIUM, 2004-2008<br />

<strong>Orphan</strong> drug<br />

(ATC<br />

code)<br />

Reimbursement<br />

application<br />

Original or<br />

revision<br />

Reimbursed Number /<br />

design<br />

Therapeutic value Therapeutic needs Budget impact<br />

Evidence<br />

published<br />

First or<br />

second<br />

line<br />

Alternative Number<br />

of<br />

patients<br />

Cost per<br />

patient per<br />

year<br />

Supplier<br />

Aldurazyme ®<br />

(A16AB05)<br />

Revision Yes 1 RCT No First No 12 € 40,000 Genzyme 1 // 1<br />

Atriance ®<br />

Original Yes 2 case series Yes Second No 5 children, € 23,000 (adult), GSK 1 // 1<br />

(L01BB07)<br />

19 adults €14,000 (child)<br />

Busilvex ®<br />

Original No 2 open-label Yes Second Yes Pierre Fabre 1 // 0<br />

(L01AB01)<br />

studies<br />

Médicament<br />

Carbaglu ®<br />

Original Yes 2 case series No First No 5-6 € 14,000- <strong>Orphan</strong> Europe 1 // 1<br />

(A16AA05)<br />

1,100,000<br />

Duodopa ®<br />

(N04BA02)<br />

Original Yes 2 case series No First Yes 80 € 41,000 Solvay Pharma 0 // 1<br />

Elaprase ®<br />

(A16AB09)<br />

Original Yes 1 RCT Yes First No 13 € 300,000 Shire 1 // 1<br />

Exjade ®<br />

Original Yes 1 RCT, 1 Yes First / Yes 450-1,150 € 12,000-23,000 Novartis pharma 1 // 1<br />

(V03AC03)<br />

case series<br />

second<br />

Fabrazyme ®<br />

Revision Yes 2 RCTs, 3 Yes First Yes 50-75 € 195,000€ Genzyme 1 // 1<br />

(A16AB04)<br />

case series<br />

Glivec ®<br />

Original No First / No € 48,000 Novartis 6 // 2<br />

(L01XE01)<br />

second<br />

Lysodren ®<br />

Original Yes Case series No First No 36 € 167,000 Laboratoire 1 // 1<br />

(L01XX23)<br />

HRA Pharma<br />

Myozyme ®<br />

Original Yes 2 open-label No First No 24 children, € 55,000- Genzyme 1 // 1<br />

(A16AB07)<br />

studies, 2<br />

case series<br />

51 adults 328,000€<br />

Nexavar ®<br />

(L01XE05)<br />

Original Yes 1 RCT Yes Second Yes 120 € 50,000 Bayer Healthcare 2 // 1<br />

Orfadin ® Original Yes Case series No First Yes 11 € 100,000 Swedish <strong>Orphan</strong> 1 // 1<br />

Number of<br />

indications<br />

(EMEA //<br />

Belgium)


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 103<br />

<strong>Orphan</strong> drug<br />

(ATC<br />

code)<br />

Reimbursement<br />

application<br />

Therapeutic value Therapeutic needs Budget impact<br />

Supplier<br />

(A16AX04) International<br />

Replagal ®<br />

(A16AB03)<br />

Revision Yes 3 case series No First Yes 50-75 € 200,000 Shire EGT 1 // 1<br />

Revatio ®<br />

Original Yes 2 RCTs Yes First / Yes 105 € 7,000-26,000 Pfizer 2 // 1<br />

(G04BE03)<br />

second<br />

Revlimid ®<br />

(L04AX04)<br />

Original Yes 2 RCTs No Second Yes 200 € 60,000 Celgene 1 // 1<br />

Savene ®<br />

(V03AF02)<br />

Original Yes 2 case series No First No 29 € 10,000 Topotarget 1 // 1<br />

Somavert ®<br />

(H01AX01)<br />

Original Yes 1 case series Yes Second No 70 € 47,000 Pfizer 1 // 1<br />

Sprycel ®<br />

(L01XE06)<br />

Original Yes 6 case series Yes Second Yes 85 € 56,000 Bristol-Myers 2 // 1<br />

Sutent ®<br />

Original Yes 1 RCT No Second Yes 73 € 16,000 Pfizer 2 // 2<br />

(L01XE04) Original Yes 2 case series Yes Second Yes 180-240 € 20,000<br />

Thelin ®<br />

Original Yes 3 RCTs No First / Yes 300 € 32,000 Encysive 2 // 2<br />

(C02KX03)<br />

second<br />

Tracleer ®<br />

Original Yes 2 RCTs No First Yes 300 € 40,000 Actelion 2 // 2<br />

(C02KX01)<br />

Registration ltd<br />

Trisenox ®<br />

(L01XX27)<br />

Original Yes 2 case series Yes Second No 9 €37,000 Cephalon 1 // 1<br />

Wilzin ®<br />

(A16AX05)<br />

Original No Yes <strong>Orphan</strong> Europe 1 // 0<br />

Xagrid ®<br />

Original Yes 6 case series Yes Second No 1,100 €8,000 Shire<br />

1 // 1<br />

(L01XX35)<br />

Pharmaceutical<br />

Zavesca ®<br />

Original Yes 1 open-label Yes Second Yes 90 € 93,000 Actelion 1 // 1<br />

(A16AV06)<br />

study, 2 case<br />

series<br />

Number of<br />

indications<br />

(EMEA //<br />

Belgium)


104 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

8.3 QUALITATIVE QUESTIONNAIRE BENCHMARKING<br />

Country Name:<br />

Name:<br />

Title:<br />

Institution:<br />

Address:<br />

Country:<br />

Telephone:<br />

Fax:<br />

Email:<br />

THE RARE DISEASE AND ORPHAN DRUG MARKET:<br />

Questionnaire<br />

Instructions: Please respond to questions electronically. You can move <strong>for</strong>ward<br />

through the main body of the questionnaire by pressing "Tab" <strong>and</strong> backwards by<br />

pressing "Shift + Tab", or you can use the scroll feature <strong>and</strong> the mouse. Boxes can be<br />

ticked <strong>and</strong> ticks can be removed by double-clicking the mouse.<br />

Identification<br />

Contact Details <strong>for</strong> the Person Completing the Form<br />

Correspondence address:<br />

Christel Fostier<br />

Yellow Window Management Consultants<br />

Lange Lozanastraat 254<br />

2018 Antwerpen<br />

Belgium<br />

T: +32/3/241.00.24<br />

F: +32/3/203.53.03<br />

E: Christel.Fostier@yellowwindow.com<br />

Section 1. Institutional context of orphan diseases/orphan drugs<br />

1.1. Identify <strong>and</strong> list centres <strong>for</strong> orphan diseases:<br />

1.2. Identify <strong>and</strong> list policy measures that promote specifically the development of orphan drugs:<br />

1.3. Identify <strong>and</strong> list specific programmes to fund research networks on orphan diseases <strong>and</strong> on orphan drugs:<br />

1.4. Identify <strong>and</strong> list incentives <strong>for</strong> research on orphan diseases <strong>and</strong> on orphan drugs:<br />

1.5. Any additional comments on the institutional context surrounding orphan diseases <strong>and</strong> orphan drugs:<br />

1.6. Are thresholds defined <strong>for</strong> reimbursement decisions <strong>for</strong> drugs expressed in<br />

QALY or similar?<br />

.a If "Yes", what is the threshold:<br />

Yes: No:


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 105<br />

.b Is this threshold also applied <strong>for</strong> orphan drugs? Or a different threshold? Yes: No:<br />

.c If a different threshold is used, which one:<br />

.d Do you expect changes in this regard in the short to medium term? Yes: No:<br />

.e If yes, please explain:<br />

1.7. Is there a national definition <strong>for</strong> “orphan disease” <strong>and</strong>/or “ultra-rare<br />

disease”?<br />

.a If “Yes”, give the definition <strong>and</strong> its source:<br />

Section 2. Marketing Authorisation of orphan drugs<br />

2.1. Is there a national procedure <strong>for</strong> granting Marketing Authorisation of<br />

orphan drugs instead of the EMEA procedure? If "No", go to<br />

question 2.2.<br />

Yes: No:<br />

Yes: No:<br />

.a If "Yes", name the organisation in charge of the national procedure <strong>for</strong> Marketing Authorisation:<br />

.b If “Yes”, specify how long it takes to obtain a Marketing Authorisation (i.e. the duration of the application<br />

procedure):<br />

.c If “Yes”, specify the various criteria that are used to judge an application:<br />

2.2. Is there a procedure <strong>for</strong> compassionate use of orphan drugs?<br />

If "No", go to question 2.3.<br />

.a If "Yes", specify the various criteria <strong>for</strong> compassionate use of orphan drugs:<br />

2.3. Is there a procedure <strong>for</strong> off-label use of orphan drugs?<br />

If "No", go to question 2.4.<br />

.a If "Yes", specify the various criteria <strong>for</strong> off-label use of orphan drugs:<br />

2.4. Any additional comments on Marketing Authorisation of orphan drugs:<br />

Yes: No:<br />

Yes: No:


106 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

Section 3. Pricing of orphan drugs<br />

3.1. Describe the mechanism by which prices of orphan drugs are set:<br />

.a Free market pricing:<br />

If "Yes", describe system of free market pricing:<br />

Yes: No:<br />

.b Fixed pricing:<br />

If "Yes", describe system of price fixing:<br />

Yes: No:<br />

.c Other (please specify): Yes: No:<br />

3.2.<br />

Describe the principal bodies or agencies that are involved in pricing of orphan drugs:<br />

3.3. Is there a procedure <strong>for</strong> revising prices of orphan drugs on<br />

national/regional lists? If "No", go to question 3.4.<br />

Yes: No:<br />

.a If "Yes", describe which factors are taken into account when revising prices (e.g. change in production<br />

costs, evolution of price index):<br />

.b If “Yes”, indicate how often prices are revised:<br />

3.4. Any additional comments on pricing system of orphan drugs:<br />

Section 4. Reimbursement of orphan drugs<br />

4.1. Describe the mechanism by which reimbursement of orphan drugs is set:<br />

.a Public procurement at national level:<br />

If "Yes", describe system of tendering:<br />

Yes: No:<br />

.b Public procurement at regional/local level:<br />

If "Yes", describe system of tendering:<br />

Yes: No:<br />

.c National list of tariffs:<br />

If "Yes", specify how tariffs are set:<br />

Yes: No:<br />

Also, specify name of national list:<br />

.d Regional list of tariffs: Yes: No:<br />

If "Yes", specify how tariffs are set:<br />

Also, specify name of regional list:<br />

.e Other (please specify): Yes: No:<br />

4.2. Which third-party payer reimburses orphan drugs?<br />

National Health Service<br />

.a (tick appropriate box)<br />

Social insurance:<br />

Public<br />

Private<br />

Combination<br />

.b Describe the process <strong>and</strong> decision criteria that are used <strong>for</strong> admitting a new orphan drug to the system of<br />

third-party payer reimbursement:<br />

.c Describe the process <strong>and</strong> decision criteria that are used <strong>for</strong> determining the level of third-party payer<br />

reimbursement of a new orphan drug:<br />

.d Are there any restrictions/conditions <strong>for</strong> reimbursement of orphan Yes: No:


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 107<br />

drugs?<br />

If "Yes", describe restrictions/conditions:<br />

.e Specify the level of patient co-payments <strong>for</strong> orphan drugs:<br />

4.3. Any additional comments on reimbursement system of orphan drugs:<br />

4.4. Is the reimbursement decision based on the EMEA dossier <strong>and</strong>/or the ICH report? If “Yes”, please describe<br />

how these documents are used:<br />

Section 5. Distribution channels<br />

5.1. Describe principal bodies or agencies that dispense orphan drugs to patients:<br />

.a Hospital pharmacies: Yes: No:<br />

.b Community pharmacies: Yes: No:<br />

.c Health authorities: Yes: No:<br />

.d Internet: Yes: No:<br />

.e Other (please specify): Yes: No:<br />

Section 6. Prescribing process<br />

6.1. Describe the mechanism by which orphan drugs are prescribed:<br />

.a Which party issues the first prescription? (tick one or more<br />

appropriate boxes)<br />

.b Are there any conditions <strong>for</strong> prescribing orphan drugs?<br />

If "No", go to question 6.2.<br />

If "Yes", describe conditions:<br />

6.2. Is there any control mechanism? Yes: No:<br />

If “Yes”, please describe this control mechanism:<br />

Specialist physician<br />

Nurse practitioner<br />

General practitioner<br />

Yes: No:<br />

6.3. Are there differences in individual reimbursement decisions depending on:<br />

Region Yes: No:<br />

<strong>Orphan</strong> drug Yes: No:<br />

Other: Yes: No:<br />

Please describe these differences:<br />

6.4. Any additional comments on prescribing process of orphan drugs:<br />

Thank you <strong>for</strong> your assistance<br />

Please return questionnaire by e-mail to:<br />

Christel.Fostier@yellowwindow.com


108 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

8.4 QUALITATIVE QUESTIONNAIRE PHARMACEUTICAL<br />

INDUSTRIES<br />

Questionnaire<br />

Study “<strong>Orphan</strong> <strong>Diseases</strong> <strong>and</strong> <strong>Orphan</strong> <strong>Drugs</strong>”<br />

Contact details <strong>for</strong> the person completing the <strong>for</strong>m<br />

Name:<br />

Title:<br />

Institution:<br />

Address:<br />

Country:<br />

Telephone:<br />

Fax:<br />

Email:<br />

Correspondence address:<br />

Christel Fostier<br />

Yellow Window Management Consultants<br />

Lange Lozanastraat 254<br />

2018 Antwerpen<br />

Belgium<br />

T: +32/3/241.00.24<br />

F: +32/3/203.53.03<br />

E: christel@yellowwindow.com<br />

1. Pros <strong>and</strong> cons of the system of orphan drug based on experience with this specific drug<br />

a. Pros:<br />

b. Cons:<br />

2. <strong>Orphan</strong> Designation<br />

a. The designation is acquired if the medical product fulfils one of the prevalence<br />

following criteria: either / or prevalence versus economic motives.<br />

Which criterion was used in this case?<br />

economic<br />

b. Has anything changed since the designation in this respect (either prevalence or economic)<br />

3. FDA versus EMEA<br />

a. Is the situation <strong>for</strong> this drug different in the USA <strong>and</strong> EU or is it<br />

likewise?<br />

b. If different, what are those differences?<br />

4. Post-Marketing Authorisation<br />

a. Have you obtained MA under exceptional circumstances or a<br />

conditional MA?<br />

i. If exceptional or conditional, under what conditions:<br />

ii. What is the actual status? (Still exceptional/conditional?)<br />

different<br />

same<br />

exceptional<br />

conditional<br />

none


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 109<br />

b. Have you set up a patient register? yes<br />

no<br />

i. Is this imposed or is it a free decision?<br />

ii. Who is managing the register?<br />

yes<br />

no<br />

5. Post-reimbursement<br />

a. Forecast of number of patients done at start versus actual number of patients<br />

identified: ……… versus ………<br />

b. Forecast budget impact versus actual budget impact: ……… versus ………<br />

c. How did you experience the interaction with the College of <strong>Orphan</strong> <strong>Drugs</strong>?<br />

d. Do all patients have access or do you notice that patients face<br />

barriers to get access?<br />

i. What types of barriers are faced?<br />

all have access<br />

barriers<br />

ii. If applicable: how many / what is the proportion of patients that do not have<br />

access? ………<br />

e. Do you consider that conditions imposed to obtain reimbursement yes<br />

are adequate?<br />

i. If not, in what sense? Has or is this changing overtime?<br />

no<br />

ii. Who plays a leading role in changing/adapting those conditions/rules?<br />

6. Present situation: could you fill in the table below <strong>for</strong> following questions<br />

a. How many patients are there in each country?<br />

b. What is the price of the drug in following countries?<br />

c. What is the turnover in each country?<br />

Belgium France Netherl<strong>and</strong>s<br />

UK Italy Sweden<br />

Number of patients<br />

Price<br />

Turnover<br />

Date of market<br />

introduction<br />

d. Do you consider all patients to have been identified or do you expect the number of<br />

patients to grow? (in Belgium)<br />

Thank you <strong>for</strong> your assistance<br />

Please return questionnaire by e-mail to:<br />

christel@yellowwindow.com


110 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

8.5 LIST OF EXPERTS AND STAKEHOLDERS CONSULTED<br />

FOR THE STUDY<br />

8.5.1 List of interview respondents<br />

Respondent Institution Date<br />

Dr Ségolène Aymé <strong>Orphan</strong>et 4/7/2008<br />

Mr André Lhoir Federal Agency <strong>for</strong> Medicines <strong>and</strong> Health Product<br />

(Belgium)<br />

12/9/2008<br />

Mr Marc Dooms UZ Leuven (Belgium) 30/9/2008<br />

Dr David Cassiman UZ Leuven (Belgium) 30/9/2008<br />

Mr Daniel Brasseur EMEA 1/10/2008<br />

Mr Erik Tambuyzer Genzyme 7/10/2008<br />

Mr Erik Brouwer<br />

Ms Katrien Van Geyt<br />

Ms Annemie Mertens<br />

Genzyme 20/10/2008<br />

Dr Jordi LLinares EMEA 14/11/2008<br />

Mr Alastair Kent Genetic Interest Group (United Kingdom) 20/11/2008<br />

Ms Françoise Marlier FPS Economy (Belgium) 12/2/2009<br />

Mr François Arickx NIHDI (Belgium) 13/2/2009<br />

Ms Minne Casteels NIHDI (Belgium) 16/2/2009<br />

Mr Michael Berntgen EMEA 18/2/009<br />

Mr Philippe Van Wilder NIHDI (Belgium) 2/4/2009<br />

Mr Paul De Keyser Independent consultant <strong>for</strong> pharmaceutical<br />

industries<br />

8.5.2 Consultations<br />

7/4/2009<br />

Consultation <strong>and</strong> exchanges took place with the Fund <strong>Rare</strong> <strong>Diseases</strong> <strong>and</strong> <strong>Orphan</strong> <strong>Drugs</strong><br />

of King Baudouin Foundation (Belgium), including participation in their meetings on the<br />

19 th of September <strong>and</strong> 14 th of October 2008.<br />

A consultation took place with Pharma.be on the 7 th of April 2009: presentation of the<br />

study <strong>and</strong> consultation on the main issues.<br />

8.5.3 National experts<br />

• France: Mrs Annie Lorence of the Afssaps<br />

• Italy: Dr Pierre Folino Gallo of the Italian Medicines Agency<br />

• Netherl<strong>and</strong>s: Dr Sonja Van Weely of the Dutch Steering Committee<br />

<strong>Orphan</strong> <strong>Drugs</strong><br />

• Sweden: Mr Karl Arnberg of the Dental <strong>and</strong> Pharmaceutical Benefits<br />

Board<br />

• United Kingdom: Ms Martina Garau of the Office of Health Economics


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 111<br />

8.6 LIST OF 14 ORPHAN DRUGS USED FOR EMEA – NIHDI<br />

COMPARISON<br />

1. Aldurazyme ®<br />

2. Atriance ®<br />

3. Elaprase ®<br />

4. Fabrazyme ®<br />

5. Nexavar ® (<strong>for</strong> indications RCC <strong>and</strong> HCC)<br />

6. Replagal ®<br />

7. Revatio ®<br />

8. Revlimid ®<br />

9. Sprycel ®<br />

10. Tasigna ®<br />

11. Tracleer ®<br />

12. Trisenox ®<br />

13. Xagrid ®<br />

14. Zavesca ®


112 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

9 REFERENCES<br />

1. Casteels M, Van Wilder P, Arickx F. De rol van de Commissie Tegemoetkoming<br />

Geneesmiddelen in het geneesmiddelenbeleid. In: Vorming verzekeringsgeneeskunde.<br />

Wetenschappelijke Vereniging voor Verzekeringsgeneeskunde; 2007.<br />

2. Drummond M, David F, Wilson A, Kanavos P, Ubel P, Rovira J. Assessing the economic<br />

challenges posed by orphan drugs. International Journal of Technology Assessment.<br />

2007;23(1):36-42.<br />

3. Van Weely S, Leufkens H. <strong>Orphan</strong> diseases. In: Priority Medicines <strong>for</strong> Europe <strong>and</strong> the World<br />

"A Public Health Approach to Innovation"; 7 October 2004. Available from:<br />

4.<br />

http://mednet3.who.int/prioritymeds/report/index.htm<br />

Veeken H, Pécoul B. <strong>Drugs</strong> <strong>for</strong> ‘neglected diseases’: a bitter pill. Tropical Medicine <strong>and</strong><br />

International Health. 2000;5(5):309-11.<br />

5. Regulation (EC) No 141/2000 of the European Parliament <strong>and</strong> the Council of 16 December<br />

1999 on orphan medicinal products, 2000.<br />

6. Australian orphan drugs policy 1997.<br />

7. Japanese <strong>Orphan</strong> Drug Regulation, 1993.<br />

8. <strong>Orphan</strong> Drug Act, 04/01/1983. Available from: http://www.fda.gov/orphan/oda.htm<br />

9. Swedish National Board of Health <strong>and</strong> Welfare. The Swedish <strong>Rare</strong> Disease Database. Available<br />

from: http://www.socialstyrelsen.se/en/rarediseases/<br />

10. Zitter M. Managing <strong>Drugs</strong> <strong>for</strong> <strong>Rare</strong> Genetic <strong>Diseases</strong>: Trends <strong>and</strong> Insights. Managed Care.<br />

February 2005:52-67.<br />

11. National Institute <strong>for</strong> Clinical Excellence. Appraising <strong>Orphan</strong> <strong>Drugs</strong>. 12/7/2005. Available from:<br />

http://www.nice.org.uk/niceMedia/pdf/smt/120705item4.pdf<br />

12. EMEA. Annual report of the European Medicines Agency 2007. In; 13 May 2008.<br />

13. Rinaldi A. Adopting an orphan. Incentives to develop drugs <strong>for</strong> rare diseases raise hopes <strong>and</strong><br />

controversy. EMBO Reports. 2005;6(6):507-10.<br />

14. Hollis A. <strong>Drugs</strong> <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong>: Paying <strong>for</strong> Innovation. In: Health Services Restructuring:<br />

New Evidence <strong>and</strong> New Directions. Kingston, Ontario; November 17-18, 2005.<br />

15. European Organisation <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong>. The Voice of 12,000 Patients. Experiences <strong>and</strong><br />

Expectations of <strong>Rare</strong> Disease Patients on Diagnosis <strong>and</strong> Care in Europe. Boulogne-Billancourt:<br />

Impression Design; 2009.<br />

16. EMEA. <strong>Orphan</strong> drugs <strong>and</strong> rare diseases at a glance. In; 3 July 2007.<br />

17. DG Enterprise <strong>and</strong> Industry, European Commission. Inventory of Community <strong>and</strong> Member<br />

States’ incentive measures to aid the research, marketing, development <strong>and</strong> availability of<br />

orphan medicinal products. Revision 2005. 07/09/2006. Available from:<br />

18.<br />

http://ec.europa.eu/enterprise/pharmaceuticals/orphanmp/doc/inventory_2006_08.pdf<br />

Haffner ME, Whitley J, Moser M. Two decades of orphan drug development. Nature Reviews<br />

Drug Discovery. 2002;1:821-5.<br />

19. Meyers A, Paola MLd. The orphan medicinal products: an international challenge. Minerva<br />

Biotecnologica. 2003;15(2):161-6.<br />

20. RIZIV. Info spot: De weesgeneesmiddelen. In; 2007.<br />

21. EBE, EuropaBio. “Towards an optimal <strong>Orphan</strong> Medicinal Products (OMP) Framework in<br />

Europe” 2005. Available from:<br />

22.<br />

http://www.maladiesrares.be/documents/<strong>Orphan</strong>s_WhitePaper.pdf<br />

Cleemput I, Neyt M, Thiry N, De Laet C, Leys M. Drempelwaarden voor kosteneffectiviteit in<br />

de gezondheidszorg. Brussel: Federaal Kenniscentrum voor de Gezondheiszorg/Centre fédéral<br />

d'expertise des soins de santé/Belgian Health Care Knowledge Centre (<strong>KCE</strong>); 2008.<br />

23. George B, Harris A, Mitchell A. Cost-effectiveness analysis <strong>and</strong> the consistency of decision<br />

making: evidence from pharmaceutical reimbursement in Australia (1991 to 1996).<br />

Pharmacoeconomics. 2001;19:1103-09.


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 113<br />

24. Clarke J. Is the current approach to reviewing new drugs condemning the victims of rare<br />

diseases to death? A call <strong>for</strong> a national orphan drug review policy. Canadian Medical<br />

Association journal. 2006;174(2):189-90.<br />

25. Owen A, Sprinks J, Meehan A, Robb T, Hardy M, Kwasha D, et al. A new model to evaluate<br />

the long-term cost effectiveness of orphan <strong>and</strong> highly specialised drugs following listing on the<br />

Australian Pharmaceutical Benefits Scheme: the Bosentan Patient Registry. Journal of Medical<br />

Economics. 2008;11(235-243).<br />

26. EMEA;c 5/8/2008. About EMEA - structure. Available from: www.emea.europa.eu<br />

27. Communication from the Commission. Guideline on aspects of the application of Article 8(2)<br />

of Regulation (EC) No 141/2000: Review of the period of market exclusivity of orphan<br />

medicinal products. In; 17.9.2008.<br />

28. Voordouw A. Het OD registratie dossier: de rol van EMEA en CBG. In: BioFarmind Seminar<br />

“Weesgeneesmiddelen, hoe pak ik het aan”. Vianen (NL), 19 April 2007; 2007.<br />

29. Llinares-Garcia J. <strong>Orphan</strong> Designation - Key Concepts <strong>and</strong> Evaluation Criteria. In. SME office;<br />

2007.<br />

30. EMEA. COMP report to the Commission in relation to article 10 of regulation 141/2000 on<br />

orphan medicinal products. European Medicines Agency; 25 July 2007. Available from:<br />

http://www.eurordis.org/article.php3?id_article=1013<br />

31. Council Regulation (EEC) No 2309/93 of 22 July 1993 laying down Community procedures <strong>for</strong><br />

the authorization <strong>and</strong> supervision of medicinal products <strong>for</strong> human <strong>and</strong> veterinary use <strong>and</strong><br />

establishing a European Agency <strong>for</strong> the Evaluation of Medicinal Products, 1993. Available from:<br />

http://ec.europa.eu/enterprise/pharmaceuticals/eudralex/vol-<br />

1/reg_1993_2309/reg_1993_2309_en.pdf<br />

32. Committee <strong>for</strong> Medicinal Products <strong>for</strong> Human Use. Guideline on clinical trials in small<br />

populations. In: EMEA, editor.; 27 July 2006.<br />

33. M<strong>and</strong>ate, Objectives <strong>and</strong> Rules of Procedure of the Scientific Advice Working Party (SAWP)<br />

2006. Available from: http://www.emea.europa.eu/pdfs/human/sciadvice/sawpm<strong>and</strong>ate.pdf<br />

34. . Fast tracking procedures <strong>and</strong> orphan drug programs in the USA, EU <strong>and</strong> Australia. Available<br />

from: http://www.findlaw.com.au/article/8338.htm<br />

35. Committee <strong>for</strong> Medicinal Products <strong>for</strong> Human Use. Dcember 2008 Plenary Meeting, Monthly<br />

Report. In: EMEA, editor.; 13 January 2009.<br />

36. Regulation (EC) No 726/2004 of the European Parliament <strong>and</strong> of the Council of 31 March 2004<br />

laying down Community procedures <strong>for</strong> the authorisation <strong>and</strong> supervision of medicinal<br />

products <strong>for</strong> human <strong>and</strong> veterinary use <strong>and</strong> establishing a European Medicines Agency, 2004.<br />

37. Haffner ME. U.S. Leads the Way in <strong>Orphan</strong> Product Legislation. In: 1st Canadian Conference<br />

on <strong>Rare</strong> Disorders & <strong>Orphan</strong> Products Policy. Ottawa, Canada; 24-25 April 2007.<br />

38. Part 316: <strong>Orphan</strong> <strong>Drugs</strong>, Revised 01/04/2008. Available from:<br />

39.<br />

http://www.accessdata.fda.gov/SCRIPTs/cdrh/cfdocs/cfcfr/CFRSearch.cfm?fr=316.3<br />

U.S. Food <strong>and</strong> Drug Administration. Office of <strong>Orphan</strong> Products Development. Available from:<br />

http://www.fda.gov/orphan/<br />

40. Karst KR. The <strong>Rare</strong>ly Used “Cost Recovery” Path to <strong>Orphan</strong> Drug Designation <strong>and</strong> Approval.<br />

FDA Law Blog. 1/2/2009.<br />

41. Mercier L, Tabutiaux-Surun A, van den Brink H, Chéron C. Les médicaments orphelins. Une<br />

harmonisation internationale à construire. Bulletin de l'ordre. 2006;393:517-26.<br />

42. FDA. How to Apply <strong>for</strong> Designation as an <strong>Orphan</strong> Product. Available from:<br />

43.<br />

http://www.fda.gov/orphan/designat/apply.htm<br />

Haffner ME. Developing <strong>Orphan</strong> Drug Products. In: 2nd International Conference on <strong>Rare</strong><br />

<strong>Diseases</strong> & <strong>Orphan</strong> <strong>Drugs</strong>. Madrid, Spain; 25/10/2006.<br />

44. Part 314: Applications <strong>for</strong> FDA approval to market a new drug Revised 01/04/2008. Available<br />

from: http://www.accessdata.fda.gov/SCRIPTs/cdrh/cfdocs/cfcfr/CFRSearch.cfm?fr=316.3<br />

45. Schuring H. Key success in orphan drug global development - an industry development. In:<br />

Genzyme Europe B.V., editor. Barcelona, 3-5 March 2008; 2008.<br />

46. Dib M, Cohen S. Traitement des maladies rares. Une législation spécifique. Neurologies.<br />

2006;9(93):654-8.


114 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

47. Organisation <strong>for</strong> Economic Co-operation <strong>and</strong> Development. Health at a glance 2007: OECD<br />

indicators. Paris: Organisation <strong>for</strong> Economic Co-operation <strong>and</strong> Development; 2007.<br />

48. Denis A, Mergaert L, Fostier C. Organisation <strong>and</strong> financing of genetic testing in Belgium. Health<br />

Services Research (HSR). Brussels: Federaal Kenniscentrum voor de Gezondheiszorg/Centre<br />

fédéral d'expertise des soins de santé/Belgian Health Care Knowledge Centre (<strong>KCE</strong>); 2007.<br />

(<strong>KCE</strong> reports 65C (D/2006/10.273/46))<br />

49. Stuurgroep Weesgeneesmiddelen. Available from: www.weesziekten.be<br />

50. Interview with Pharmacist Francis Arickx. 13/2/2009.<br />

51. Ministerieel besluit betreffende de prijzen van de terugbetaalbare geneesmiddelen, 29/12/1989.<br />

52. Koninklijk Besluit van 21 december 2001 tot vaststelling van de procedures, termijnen en<br />

voorwaarden inzake de tegemoetkoming van de verplichte verzekering voor geneeskundige<br />

verzorging en uitkeringen in de kosten van farmaceutische specialiteiten, 21/09/2002.<br />

53. DG Enterprise EC;c 11/3/2009. Register of designated <strong>Orphan</strong> Medicinal Products. Available<br />

from: http://ec.europa.eu/enterprise/pharmaceuticals/register/orphreg.htm<br />

54. Wet houdende herziening van de farmaceutische wetgeving, 1/5/2006.<br />

55. Trouet C, Gobert M, Podoor M. Clinical Trials in Belgium: The Belgian implementation of the<br />

European Clinical Trials Directives. An operational guide. 2nd edition ed. Antwerp-Ox<strong>for</strong>d:<br />

Intersentia; 2007.<br />

56. Wet van 27/4/2005 betreffende de beheersing van de begroting van de gezondheidszorg en<br />

houdende diverse bepalingen inzake gezondheid 20/5/2005.<br />

57. Bogaert M. De terugbetaling van weesgeneesmiddelen in België. In: Weesgeneesmiddelen in<br />

België Brussels; 7 December 2006.<br />

58. GIS - Institut des Maladies <strong>Rare</strong>s. Available from: http://www.institutmaladiesrares.net/<br />

59. Ministère de la Santé, de la Jeunesse, des Sports, et de la Vie Associative;c March 2004. Les<br />

médicaments orphelins. Available from:<br />

60.<br />

www.sante.gouv.fr/htm/dossiers/maladies_rares/maladies_6bis.htm<br />

<strong>Rare</strong> <strong>Diseases</strong> Task Force. Centres of Reference <strong>for</strong> rare diseases in Europe: State-of-the-art in<br />

2006 <strong>and</strong> recommendations of the <strong>Rare</strong> <strong>Diseases</strong> Task Force. December 2006. Available from:<br />

http://ec.europa.eu/health/ph_threats/non_com/docs/contribution_policy.pdf<br />

61. Ministère de la Santé, de la Jeunesse, des Sports, et de la Vie Associative;c 18/2/2009. Les<br />

Maladies Orphelines. Available from: http://www.sante-sports.gouv.fr/dossiers/sante/maladiesrares/maladies-rares.html?var_recherche=orphelin<br />

62. Lorence A. Les médicaments orphelins en Europe. In: Afssaps, editor. MEDEC; 15/03/2006.<br />

63. Meyer F. <strong>Orphan</strong> <strong>Drugs</strong>: How Are They Assessed / Appraised in France? In: HAS, editor. 25<br />

February 2008.<br />

64. Meyer F. Assessment, Pricing of Pharmaceuticals in France. In: AIFA. Rome; March 2007.<br />

65. Ministère de la Santé, de la Jeunesse, des Sports, et de la Vie Associative. Nouvelle organisation<br />

/ L'Union des Caisses d'Assurance Maladie (UNCAM). Available from:<br />

66.<br />

http://www.sante.gouv.fr/assurance_maladie/organisation/uncam.htm<br />

Bélorgey C. Mise à disposition précoce des médicaments orphelins en France In: Journées de<br />

l'Afssaps.<br />

67. Les entreprises du médicament. Haute Aurorité de Santé et Commission de Transparence.<br />

Available<br />

http://www.leem.org/htm/themes/imprimer.asp?id_sous_rubrique=91&id_article=318<br />

from:<br />

68. Seget S. Pharmaceutical Pricing Strategies: Optimizing returns throughout R&D <strong>and</strong> marketing.<br />

Reuters Business Insight Healthcare; 2003.<br />

69. Folino Gallo P. Benchmarking questionnaire Italy. 16/3/2009.<br />

70. . National Centre <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong>. Available from: http://www.iss.it/cnmr<br />

71. Taruscio D. Italian Network <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong>: legislation <strong>and</strong> organization. National Centre<br />

<strong>Rare</strong> <strong>Diseases</strong> Istituto Superiore di Sanità Italy. Available from:<br />

http://www.orpha.net/testor/doc/rdtf/wg/italy.pdf


<strong>KCE</strong> Reports 112 <strong>Orphan</strong> <strong>Drugs</strong> 115<br />

72. Folino Gallo P. <strong>Orphan</strong> <strong>Drugs</strong> in Italy. Accommodating orphan drugs: balancing innovation <strong>and</strong><br />

financial stability. In: Accommodating orphan drugs: balancing innovation <strong>and</strong> financial stability.<br />

London; 25 February 2008.<br />

73. International Society <strong>for</strong> Pharmacoeconomics <strong>and</strong> Outcomes Research. Reimbursement<br />

Processes Around the World: Italy. October 2008. ISPOR Global Health Care Systems Road<br />

Map Available from: http://www.ispor.org/HTARoadMaps/Italy.asp<br />

74. Van Weely S. Benchmarking questionnaire <strong>for</strong> the Netherl<strong>and</strong>s. 2009.<br />

75. Heemstra HE, Vrueh Rd, Weely Sv, Leufkens HABH. <strong>Orphan</strong> drug development across<br />

Europe: bottlenecks <strong>and</strong> opportunities. Drug Discovery Today. June 2008.<br />

76. College ter Beoordeling van Geneesmiddelen. Missie, visie en ambitie. Available from:<br />

http://www.cbg-meb.nl/CBG/nl/over-ons/organisatie/Missie_Visie_Ambitie/default.htm<br />

77. College ter Beoordeling van Geneesmiddelen. Compassionate use. Available from:<br />

http://www.cbg-meb.nl/CBG/nl/humane-geneesmiddelen/registratiezaken/compassionate-useprogramma/default.htm<br />

78. College ter Beoordeling van Geneesmiddelen. Off-label use. Available from: http://www.cbgmeb.nl/CBG/nl/humane-geneesmiddelen/geneesmiddelen/Off-label-use/default.htm<br />

79. Regeling zorgverzekering, 1 september 2005. Available from: http://www.st-<br />

80.<br />

ab.nl/wetzvwor1rz.htm#h2p2<br />

College voor Zorgverzekeringen, Ministerie van Volksgezondheid, Welzijn en Sport. Procedure<br />

beoordeling extramurale geneesmiddelen. 27016438 Available from:<br />

http://www.cvz.nl/resources/CFH_procedure%20beoord%20extrm%20geneesm_tcm28-<br />

15809.pdf<br />

81. Dental <strong>and</strong> Pharmaceutical Benefits Agency. Welcome to TLV. Available from:<br />

82.<br />

http://www.tlv.se/in-english/<br />

Ramsberg J. Accomodating orphan drugs in Sweden. In: Board PB, editor. Accommodating<br />

orphan drugs: balancing innovation <strong>and</strong> financial stability. London, 25 February 2008; 2008.<br />

83. Moïse P, Docteur E. Pharmaceutical Pricing <strong>and</strong> Reimbursement <strong>Policies</strong> in Sweden. OECD;<br />

2007. OECD Health Working Papers No. 28 Available from:<br />

84.<br />

http://titania.sourceoecd.org/vl=2094225/cl=25/nw=1/rpsv/cgi-bin/wppdf?file=5l4jn1vgr1kc.pdf<br />

Arnberg K. Benchmarking questionnaire <strong>for</strong> Sweden. 12/3/2009.<br />

85. Pharmaceutical Benefits Board. General guidelines concerning price increases of<br />

pharmaceuticals from the Pharmaceutical Benefits Board 2006. Available from:<br />

86.<br />

http://www.tlv.se/Upload/English/ENG-lfnar-2006-1.pdf<br />

Garau M. Benchmarking questionnaire <strong>for</strong> the United Kingdom. 2009.<br />

87. National Institute <strong>for</strong> Clinical Excellence. Appraising life-extending, end of life treatments. In;<br />

2009.<br />

88. Rawlins MD, Culyer AJ. National Institute <strong>for</strong> Clinical Excellence <strong>and</strong> its value judgments. BMJ<br />

(Clinical research ed.). 2004(329):224-7.<br />

89. Smith R. Nice decisions on drugs are flawed <strong>and</strong> tossing a coin is fairer, says academic.<br />

Telegraph 23/10/2008.<br />

90. National Commissioning Group Available from: http://www.ncg.nhs.uk/faqs.htm<br />

91. Medicines <strong>and</strong> Healthcare products Regulatory Agency. Medicines that do not need a licence<br />

(Exemptions from licensing). Available from:<br />

92.<br />

http://www.mhra.gov.uk/Howweregulate/Medicines/Doesmyproductneedalicence/Medicinesthat<br />

donotneedalicence/CON009278<br />

McCabe C, Claxton K, Tsuchiya A. <strong>Orphan</strong> drugs <strong>and</strong> the NHS: Should we value rarity? British<br />

Medical Journal 2006;99(5):341-5.<br />

93. National Institute <strong>for</strong> Clinical Excellence. Guide to the Methods of Technology Appraisal. In;<br />

April 2004.<br />

94. Garau M, Mestre-Ferr<strong>and</strong>iz J. European medicines pricing <strong>and</strong> reimbursment. Now <strong>and</strong> the<br />

future. Office of Health Economics; 2009.<br />

95. AWMSG;c 2008. Appraisals. Available from:<br />

http://www.wales.nhs.uk/sites3/page.cfm?orgid=371&pid=9323


116 <strong>Orphan</strong> <strong>Drugs</strong> <strong>KCE</strong> Reports 112<br />

96. Garau M, Chauhan D. Decision-making processes in the orphan drugs arena. The UK<br />

perspective. In: Office of Health Economics, editor. Accommodating orphan drugs: balancing<br />

innovation <strong>and</strong> financial stability. London; 25 February 2008.<br />

97. Scottish Medicines Consortium [cited 05/12/2008]. <strong>Orphan</strong> <strong>Drugs</strong> (December 2007). Available<br />

from: http://www.scottishmedicines.org.uk/smc/3869.html<br />

98. Burls A. Ethical issues in orphan drugs - finding a way <strong>for</strong>ward. In: Evidence-Based Health Care<br />

Symposium. Jagiellonian University, Krakow; 2007.<br />

99. Garau M, Mestre-Ferr<strong>and</strong>iz J, Wang Q. Access Mechanisms <strong>for</strong> <strong>Orphan</strong> <strong>Drugs</strong>: A Comparative<br />

Study of Selected European Countries. London: Office of Health Economics; Forthcoming.<br />

100. Department of Health. The Pharmaceutical Price Regulation Scheme. 2009. Available from:<br />

http://www.dh.gov.uk/en/Publications<strong>and</strong>statistics/Publications/DH_091825<br />

101. WHO Collaborating Centre <strong>for</strong> Drug Statistics Methodology. Anatomical Therapeutic<br />

Chemical classification system <strong>for</strong> drugs. Available from: http://www.whocc.no<br />

102. RIZIV;c 1/3/2009. Farmaceutische specialiteiten. Available from:<br />

http://www.riziv.fgov.be/inami_prd/ssp/cns2/pages/SpecialityCns.asp<br />

103. RIZIV (Dienst voor Geneeskundige Verzorging). Monitoring Of Reimbursement Significant<br />

Expenses. M.O.R.S.E. semesterieel rapport 2008 (1) gegevens 1e semester 2008. RIZIV; 2008.<br />

Available from: http://www.riziv.fgov.be/drug/nl/statistics-scientificin<strong>for</strong>mation/report/pdf/morse.pdf<br />

104. European Organisation <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong>. EURORDIS Position Paper on the “Centralised<br />

procedure <strong>for</strong> the scientific assessment of the Therapeutic Added Value of <strong>Orphan</strong> <strong>Drugs</strong>”.<br />

February 2008. Available from: http://www.eurordis.org/IMG/pdf/position-paper-EURORDIStherapeutic-added-value-ODFeb08.pdf<br />

105. Working Group Pricing <strong>and</strong> Reimbursement. Improving access to orphan medicines <strong>for</strong> all<br />

affected EU citizens. In: The Pharmaceutical Forum, editor.; 2008.<br />

106. Connock M, Juarez-Garcia A, Frew E, Mans A, Dretzke J, Fry-Smith A, et al. A systematic<br />

review of the clinical effectiveness <strong>and</strong> cost-effectiveness of enzyme replacement therapies <strong>for</strong><br />

Fabry’s disease <strong>and</strong> mucopolysaccharidosis type I. Health Technology Assessment. 2006;10(20).


This page is left intentionally blank.


Legal depot : D/2009/10.273/32


<strong>KCE</strong> reports<br />

33 Effects <strong>and</strong> costs of pneumococcal conjugate vaccination of Belgian children. D/2006/10.273/54.<br />

34 Trastuzumab in Early Stage Breast Cancer. D/2006/10.273/25.<br />

36 Pharmacological <strong>and</strong> surgical treatment of obesity. Residential care <strong>for</strong> severely obese children<br />

in Belgium. D/2006/10.273/30.<br />

37 Magnetic Resonance Imaging. D/2006/10.273/34.<br />

38 Cervical Cancer Screening <strong>and</strong> Human Papillomavirus (HPV) Testing D/2006/10.273/37.<br />

40 Functional status of the patient: a potential tool <strong>for</strong> the reimbursement of physiotherapy in<br />

Belgium? D/2006/10.273/53.<br />

47 Medication use in rest <strong>and</strong> nursing homes in Belgium. D/2006/10.273/70.<br />

48 Chronic low back pain. D/2006/10.273.71.<br />

49 Antiviral agents in seasonal <strong>and</strong> p<strong>and</strong>emic influenza. Literature study <strong>and</strong> development of<br />

practice guidelines. D/2006/10.273/67.<br />

54 Cost-effectiveness analysis of rotavirus vaccination of Belgian infants D/2007/10.273/11.<br />

59 Laboratory tests in general practice D/2007/10.273/26.<br />

60 Pulmonary Function Tests in Adults D/2007/10.273/29.<br />

64 HPV Vaccination <strong>for</strong> the Prevention of Cervical Cancer in Belgium: Health Technology<br />

Assessment. D/2007/10.273/43.<br />

65 Organisation <strong>and</strong> financing of genetic testing in Belgium. D/2007/10.273/46.<br />

66. Health Technology Assessment: Drug-Eluting Stents in Belgium. D/2007/10.273/49.<br />

70. Comparative study of hospital accreditation programs in Europe. D/2008/10.273/03<br />

71. Guidance <strong>for</strong> the use of ophthalmic tests in clinical practice. D/200810.273/06.<br />

72. Physician work<strong>for</strong>ce supply in Belgium. Current situation <strong>and</strong> challenges. D/2008/10.273/09.<br />

74 Hyperbaric Oxygen Therapy: a Rapid Assessment. D/2008/10.273/15.<br />

76. Quality improvement in general practice in Belgium: status quo or quo vadis?<br />

D/2008/10.273/20<br />

82. 64-Slice computed tomography imaging of coronary arteries in patients suspected <strong>for</strong> coronary<br />

artery disease. D/2008/10.273/42<br />

83. International comparison of reimbursement principles <strong>and</strong> legal aspects of plastic surgery.<br />

D/200810.273/45<br />

87. Consumption of physiotherapy <strong>and</strong> physical <strong>and</strong> rehabilitation medicine in Belgium.<br />

D/2008/10.273/56<br />

90. Making general practice attractive: encouraging GP attraction <strong>and</strong> retention D/2008/10.273/66.<br />

91 Hearing aids in Belgium: health technology assessment. D/2008/10.273/69.<br />

92. Nosocomial Infections in Belgium, part I: national prevalence study. D/2008/10.273/72.<br />

93. Detection of adverse events in administrative databases. D/2008/10.273/75.<br />

95. Percutaneous heart valve implantation in congenital <strong>and</strong> degenerative valve disease. A rapid<br />

Health Technology Assessment. D/2008/10.273/81<br />

100. Threshold values <strong>for</strong> cost-effectiveness in health care. D/2008/10.273/96<br />

102. Nosocomial Infections in Belgium: Part II, Impact on Mortality <strong>and</strong> Costs. D/2009/10.273/03<br />

103 Mental health care re<strong>for</strong>ms: evaluation research of ‘therapeutic projects’ - first intermediate<br />

report. D/2009/10.273/06.<br />

104. Robot-assisted surgery: health technology assessment. D/2009/10.273/09<br />

108. Tiotropium in the Treatment of Chronic Obstructive Pulmonary Disease: Health Technology<br />

Assessment. D/2009/10.273/20<br />

109. The value of EEG <strong>and</strong> evoked potentials in clinical practice. D/2009/10.273/23<br />

111. Pharmaceutical <strong>and</strong> non-pharmaceutical interventions <strong>for</strong> Alzheimer’s Disease, a rapid<br />

assessment. D/2009/10.273/29<br />

112 <strong>Policies</strong> <strong>for</strong> <strong>Rare</strong> <strong>Diseases</strong> <strong>and</strong> <strong>Orphan</strong> <strong>Drugs</strong>. D/2009/10.273/32.<br />

This list only includes those <strong>KCE</strong> reports <strong>for</strong> which a full English version is available. However, all <strong>KCE</strong><br />

reports are available with a French or Dutch executive summary <strong>and</strong> often contain a scientific<br />

summary in English.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!